WO2022173333A2 - Composés, compositions et méthodes de traitement de maladies et d'états liés à l'âge - Google Patents

Composés, compositions et méthodes de traitement de maladies et d'états liés à l'âge Download PDF

Info

Publication number
WO2022173333A2
WO2022173333A2 PCT/RU2022/050044 RU2022050044W WO2022173333A2 WO 2022173333 A2 WO2022173333 A2 WO 2022173333A2 RU 2022050044 W RU2022050044 W RU 2022050044W WO 2022173333 A2 WO2022173333 A2 WO 2022173333A2
Authority
WO
WIPO (PCT)
Prior art keywords
days
disease
compound selected
treatment
subject
Prior art date
Application number
PCT/RU2022/050044
Other languages
English (en)
Inventor
Andrei Evgenevich TARKHOV
Konstantin Aleksandrovich AVKHACHEV
Olga Andreevna BURMISTROVA
Maksim Nikolaevich KHOLIN
Petr Olegovich Fedichev
Original Assignee
Gero Pte. Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gero Pte. Ltd. filed Critical Gero Pte. Ltd.
Priority to EP22753056.5A priority Critical patent/EP4291239A2/fr
Publication of WO2022173333A2 publication Critical patent/WO2022173333A2/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • COMPOUNDS, COMPOSITIONS AND METHODS FOR TREATING AGE-RELATED DISEASES AND CONDITIONS FIELD [0001] We disclose the anti-aging, senolytic and other therapeutic effects of several compounds and their analogs and combinations described herein as well as related methods. BACKGROUND [0002] The development of methods, medications for increasing health span and/or treating aging or age-related diseases would benefit many. [0003] New targets, compounds, compositions for treating aging or age-related conditions are desired. SUMMARY [0004] The present inventors identified therapeutic effects of several compounds and their analogs and combinations described herein as well as related methods.
  • the present application includes new anti-aging and other therapeutic uses disclosed herein of modulators of target selected from the group of targets consisting of: , S C6 3 [1]
  • the term “Target” means any one of the proteins selected from the group consisting of: [2]
  • the term “Targets” means all of the proteins selected from the group consisting of:
  • Targets means any combination of the proteins selected from the group consisting of:
  • modulators of any one of the Targets are inhibitors. In some embodiments, modulators of Targets are small molecules. In some embodiments, modulators of any one of the Targets are small molecule inhibitors. In some embodiments such modulators of any one of the
  • Targets are selected from Table Table 1 ah and Table 1c or their pharmaceutically acceptable salt or any combination of it or compounds or, optionally, having the similar SAR characteristics salts and/or solvates of any one of inhibitors of any of the Targets.
  • the anti-aging compound binds and/or inhibits at least one of the
  • this invention is a method of anti-aging treatmemt, comprising adminestring of inhibitor of Target in therapeutically effective amount.
  • the application also provides an anti-aging therapy, comprising a compound selected from the group of all compounds of Table 1 ah and Table 1c .
  • the application provides an anti-aging therapy, comprising an inhibitor of the protein, wherein protein is selected from the group consisting of: , , , , ,
  • the application provides an anti-aging therapy, comprising an inhibitor of the gene, wherein gene is selected from the group consisting of: , , , , ,
  • the application provides an anti-aging therapy, comprising a compound selected from the group consisting of:
  • the application also provides an anti-aging therapy, comprising a compound selected from the group of all compounds of Table 1 ab and Table 1c .
  • the application also provides an senolytic therapy, comprising a compound selected from the group of all compounds of Table 1 ab and Table 1c .
  • the application also provides an senostatic therapy, comprising a compound selected from the group of all compounds of Table 1 ab and Table 1c .
  • the application also provides adjuvant tumour therapy, comprising a compound selected from the group of all compounds of Table 1 ab and Table 1c .
  • the application also provides an anti-aging therapy, comprising the compound selected from Table 1 ab and Table 1c and an enhancer moiety.
  • the enhancer moiety is a permeability enhancer, stability enhancer or bioavailability enhancer.
  • the application also provides a composition comprising a compound as described herein or a compound as described herein; and a carrier.
  • the application also provides a pharmaceutical composition comprising a compound as described herein; and a pharmaceutically acceptable carrier.
  • any one of the inhibitors of any one of the Targets including but not limited to any one of the compounds selected from Table 1 ab and Table 1c has at least one of the effects selected from the following group consisting of: ameliorates at least one symptom of the age-related disease or condition in the subject, delays mortality, delays onset of at least one aging associated diseases, decreases frailty, e.g. the Frailty Index (e.g.
  • the term “anti-aging” is used interchangeably with the term “ senolytic”.
  • the term “anti-aging therapy” is used interchangeably with the term “Senostatic”.
  • the application includes a use of a compound selected from Table 1 ab and Table 1c for treating or preventing of the condition selected from the group consisting of: aging, frailty, an age-related disease or condition in a subject; or for delaying or reversing one or more signs or symptoms of aging in a subject and/or for increasing longevity in a subject.
  • the application also provides a use of the inhibitor of Target, including but not limited to any one compound selected from Table 1 ab and Table 1c for treating or preventing a senescence associated disease or disorder in a subject and/or for selectively killing one or more senescent cells in a subject.
  • the application further provides a use of the compound selected from Table 1 ab and Table 1c for treating or preventing viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject or for alleviating signs and symptoms of viral disease, optionally COVID-19 or influenza, or infectious disease in a subject.
  • the anti-aging drug is a molecule that binds any at least one of the Targets or/and inhibits activity of such Target.
  • the anti-aging therapy includes the compound selected from the Table 1 ab and Table 1c conjugated to an enhancer moiety or a composition comprising the compound or compound or an antibody or antibody fragment that binds target selected from the group of Targets.
  • the age-related disease or condition is selected from the group consisting of age-related tissue decline, age-related organ decline, degenerative disease, function- decreasing disorder, cancer, Alzheimer's disease, Parkinson's disease, cataracts, macular degeneration, glaucoma, atherosclerosis, acute coronary syndrome, myocardial infarction, stroke, recovery after stroke, PSD-95 decrease, hypertension, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), type 1 diabetes, obesity, fat dysfunction, coronary artery disease, cerebrovascular disease, periodontal disease, cancer treatment-related disability, chemotherapy treatment-related disability, chemotherapy treatment-related frailty, frailty, radiotherapy and other radiation related disability, chemotherapy treatment-related frailty, cancer treatment-related atrophy, cancer treatment-related fibrosis, brain injury, heart injury, and therapy-related myelodysplastic syndrome, accelerated aging, accelerated aging disease, Hutchinson-Gilford progeria syndrome
  • the subject is a mammal, optionally a human, optionally a human of at least 60, 65, 70, 75, 80, 85, 90, 95 or 100 years of age.
  • the wording “the compound selected from Table 1 ab and Table 1c ” means “the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c ”.
  • the wording “the compound selected from Table 1 ab and Table 1c ” or “the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c ” means “the pharmaceutical composition selected from the group consisting of all pharmaceutical compositions listed in the Table 1 ab and Table 1c ”. [0029] In some embodiments, the wording “the compound selected from Table 1 ab and Table 1c ” means compound selected from the group, consisting of:
  • the wording “the compound selected from Table 1 ab and Table 1c ” or “the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c ” means Active pharmaceutical ingredient (API), selected from the group consisting of all Active pharmaceutical ingredients of all of the drugs, drug candidates, pharmaceutical compositions and other agents listed in the Table 1 ab and Table 1c .
  • API Active pharmaceutical ingredient
  • the wordings “the compound selected from Table 1 ab and Table 1c ”, “the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c ”, “the pharmaceutical composition selected from the group consisting of all pharmaceutical compositions listed in the Table 1 ab and Table 1c ” and alike can be used interchangebly with the wording “the agent reducing, inhibiting, or degrading a protein selected from the group consisting of: [0032] In some embodiments, compound selected from the table 1 ab and Table 1c is used for Simultaneous Alleviation of Co-morbidities.
  • a compound selected from the table 1 ab and Table 1c alleviates one or more of: mild diabetes, atherosclerosis, hypertension, mild cognitive impairment, sarcopenia, osteoarthritis, mild renal insufficiency, etc. occurring within a subject.
  • a compound selected from the table 1 ab and Table 1c is used for improving parameter selected from the group consisting of: glucose tolerance, carotid flow velocity, blood pressure, timed walking ability, walking speed, joint pain, joint pain inventories, creatinine clearance.
  • compound selected from the table 1 ab and Table 1c is used for Delaying Accelerated Aging-like Conditions.
  • compound selected from the table 1 ab and Table 1c is used for one of the selected from the group: treatment of the frailty, improving of at least one of endurance parametered, improving of at least one of metabolic parameters, improving of at least one of cardiovascular functions, improving of at least one of cognitive functions, alleviating at least one symptome in childhood cancer survivors, alleviating at least one symptome caused by chemothrepy in childhood cancer survivors, alleviating at least one symptome caused by chemotherapy, alleviating at least one symptome caused by radiotherapy, alleviating at least one symptome in bone marrow transplant survivors, alleviating at least one symptome casued by bone marrow transplantion, alleviating at least one progeroid syndrome, patients alleviating at least one symptome of diabetes due to obesity, alleviating at least one symptome of diabetes, alleviating at least one symptome of HIV or AIDS, alleviating at least one of conditions related to latent viruses, such as HIV.
  • compound selected from the table 1 ab and Table 1c is used for treating disease selected from the group consisting of: osteoarthritis, fracture non-union, glaucoma and macular degeneration, sites subjected to therapeutic radiation, the lungs in idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, or damage due to tobacco, or atherosclerotic plaques (by catheterization), among others.
  • disease selected from the group consisting of: osteoarthritis, fracture non-union, glaucoma and macular degeneration, sites subjected to therapeutic radiation, the lungs in idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, or damage due to tobacco, or atherosclerotic plaques (by catheterization), among others.
  • disease selected from the group consisting of: osteoarthritis, fracture non-union, glaucoma and macular degeneration, sites subjected to therapeutic radiation, the lungs in idiopathic pulmonary fibrosis, chronic obstructive
  • compound selected from the table 1 ab and Table 1c is used for increasing Resilience or Clinical Stresses in Pre-frail Subjects. Capacity to recover following a medical or physiological stress generally declines with aging.
  • compound selected from the table 1 ab and Table 1c is used to increase resilience. An example might include the use of such agents before chemotherapy in an effort to improve recovery and to allow providing higher, more effective doses of chemotherapy to elderly frail subjects with high initial burdens of senescent cells.
  • compound selected from the table 1 ab and Table 1c is used for targeting basic aging processes, to accelerate recovery after elective surgery, bone marrow transplantation, therapeutic radiation, pneumonia, or myocardial infarction or to enhance immune response to influenza vaccination.
  • compound selected from the table 1 ab and Table 1c is used for two weeks before influenza vaccination results in better influenza antibody generation in elderly subjects (for example, the same protocol as described Mannick et al., 2014). In some embodiments, compound selected from the table 1 ab and Table 1c administered as a single course before vaccination is used for the same. [0038] In some embodiments, compound selected from the table 1 ab and Table 1c is used for reducing frailty, including use to alleviate slow gait, decreased strength, or sarcopenia or delay loss of independence.
  • compound selected from the table 1 ab and Table 1c is used for reducing frailty in progeroid patients
  • a method is provided for treating, reducing the likelihood of occurrence of, or delaying onset of a senescent cell-associated disease or disorder in a subject who has a senescent cell-associated disease or disorder or who has at least one predisposing factor for developing the senescent cell-associated disease or disorder, comprising administering to the subject a compound selected from the table 1 ab and Table 1c that alters either one or both of a cell survival signaling pathway and an inflammatory pathway in the senescent cell, thereby promoting death of the senescent cell, with the proviso that if the subject has a cancer, the compound selected from the table 1 ab and Table 1c is not a primary therapy for treating the cancer, wherein the compound selected from the table 1 ab and Table 1c is administered once every 0.5-12 months, and wherein the senescent cell-associated disease or disorder is a cardiovascular
  • a method for treating, reducing the likelihood of occurrence of, or delaying onset of a senescent cell-associated disease or disorder in a subject who has a senescent cell-associated disease or disorder or who has at least one predisposing factor for developing the senescent cell-associated disease or disorder comprising administering to the subject a compound selected from the table 1 ab and Table 1c that alters either one or both of a cell survival signaling pathway and an inflammatory pathway in the senescent cell, thereby promoting death of the senescent cell, wherein the compound selected from the table 1 ab and Table 1c is administered once every 4-12 months.
  • the treatment regimen of the methods for treating a senescence associated disease or disorder comprises administering a compound selected from the table 1 ab and Table 1c for a time sufficient and in an amount sufficient that selectively kills senescent cells.
  • the compound selected from the table 1 ab and Table 1c is administered within a treatment cycle, which treatment cycle comprises a treatment course followed by a non-treatment interval.
  • a treatment course of administration refers herein to a finite time frame over which one or more doses of the compound selected from the table 1 ab and Table 1c on one or more days are administered.
  • the finite time frame may be also called herein a treatment window.
  • a method for treating a senescence-associated disease or disorder, which is not a cancer, and which method comprises administering to a subject in need thereof a small molecule compound selected from the table 1 ab and Table 1c that selectively kills senescent cells and is administered within a treatment cycle.
  • the methods comprise administering the compound selected from the table 1 ab and Table 1c in at least two treatment cycles.
  • the non-treatment interval may be at least about 2 weeks or between at least about 0.5-12 months, such as at least about one month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, or at least about 12 months (i.e., 1 year).
  • the non-treatment interval is between 1-2 years or between 1-3 years, or longer.
  • each treatment course is no longer than about 1 month, no longer than about 2 months, or no longer than about 3 months; or is no longer than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 26, 27, 28, 29, 30, or 31 days.
  • the treatment window i.e., treatment course
  • a single treatment course occurs over no longer than 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 26, 27, 28, 29, 30, or 31 days.
  • the compound selected from the table 1 ab and Table 1c may be administered at least on two days (i.e., two days or more) with a variable number of days on which the agent is not administered between the at least two days of administration.
  • the treatment course may have one or more intervals of one or more days when the Compound selected from the table 1 ab and Table 1c , is not administered.
  • the agent when the compound selected from the table 1 ab and Table 1c is administered on 2 or more days during a treatment course not to exceed 21 days, the agent may be administered on any total number of days between from 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 26, 27, 28, 29, 30, or 31 days.
  • the compound selected from the table 1 ab and Table 1c is administered to a subject during a treatment course of 3 days or more, and the agent may be administered every 2 nd day (i.e., every other day).
  • the compound selected from the table 1 ab and Table 1c when the compound selected from the table 1 ab and Table 1c is administered to a subject for a treatment window of 4 days or more, the compound selected from the table 1 ab and Table 1c may be administered every 3 rd day (i.e., every other third day).
  • the compound selected from the table 1 ab and Table 1c is administered on at least two days (i.e., 2 or more) during a treatment course that is at least 2 days and no more than about 21 days (i.e., from about 2-21 days); at least 2 days and no longer than about 14 days (i.e., from about 2-14 days); at least 2 days and no longer than about 10 days (i.e., from about 2-10 days); or at least 2 days and no longer than about 9 days (i.e., from about 2-9 days); or at least 2 days and no longer than about 8 days (i.e., from about 2-8 days).
  • the compound selected from the table 1 ab and Table 1c is administered on at least two days (i.e., 2 or more) during a treatment window is at least 2 days and no longer than about 7 days (i.e., from about 2-7 days); at least 2 days and no longer than about 6 days (i.e., from about 2-6 days) or at least 2 days and no more than about 5 days (i.e., from about 2-5 days) or at least 2 days and no longer than about 4 days (i.e., from about 2-4 days).
  • the treatment window is at least 2 days and no longer than 3 days (i.e., 2-3 days), or 2 days. In certain particular embodiments, the treatment course is no longer than 3 days.
  • the treatment course is no longer than 5 days. In still other specific embodiments, the treatment course is no longer than 7 days, 10 days, or 14 days or 21 days.
  • the compound selected from the table 1 ab and Table 1c is administered on at least two days (i.e., 2 or more days) during a treatment window that is at least 2 days and no longer than about 11 days (i.e., 2-11 days); or the compound selected from the table 1 ab and Table 1c is administered on at least two days (i.e., 2 or more days) during a treatment window that is at least 2 days and no longer than about 12 days (i.e., 2-12 days); or the compound selected from the table 1 ab and Table 1c is administered on at least two days (i.e., 2 or more days) during a treatment window that is at least 2 days and no more than about 13 days (i.e., 2-13 days); or the compound selected from the table 1 ab and Table 1c is administered on at least two days (i.e., 2 or more days) during
  • the compound selected from the table 1 ab and Table 1c may be administered on at least 3 days over a treatment course of at least 3 days and no longer than any number of days between 3 and 21 days; or is administered on at least 4 days over a treatment course of at least 4 days and no longer than any number of days between 4 and 21 days; or is administered on at least 5 days over a treatment course of at least 5 days and no longer than any number of days between 5 and 21 days; or is administered on at least 6 days over a treatment course of at least 6 days and no longer than any number of days between 6 and 21 days; or is administered at least 7 days over a treatment course of at least 7 days and no longer than any number of days between 7 and 21 days; or is administered at least 8 or 9 days over a treatment course of at least 8 or 9 days, respectively, and no longer than any number of days between 8 or 9 days, respectively, and 21 days; or is administered at least 10 days over a treatment course of at least 10 days and no longer than any number of days between 10 and 21 days; or is administered at least 10 days over
  • a compound selected from the table 1 ab and Table 1c may be administered on at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, and 14 days over a treatment of window of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, and 14 days, respectively, and no longer than 14 days.
  • a compound selected from the table 1 ab and Table 1c may be administered on at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 days over a treatment of window of at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 days, respectively, and no longer than 10 days.
  • a compound selected from the table 1 ab and Table 1c may be administered on at least 2, 3, 4, 5, 6, or 7 days over a treatment window of at least 2, 3, 4, 5, 6, or 7 days, respectively, and no longer than 7 days.
  • a compound selected from the table 1 ab and Table 1c may be administered on at least 2, 3, 4, or 5 days over a treatment of window of at least 2, 3, 4, or 5 days, respectively, and no longer than 5 days.
  • doses of the compound selected from the table 1 ab and Table 1c may be administered for a lesser number of days than the total number of days within the particular treatment window.
  • the number of days on which the compound selected from the table 1 ab and Table 1c may be administered is any number of days between 2 days and 7, 10, 14, or 21 days, respectively, and at any interval appropriate for the particular disease being treated, the compound selected from the table 1 ab and Table 1c being administered, the health status of the patient and other relevant factors, which are discussed in greater detail herein.
  • the agent may be delivered on the minimum number days of the window, the maximum number of days of the window, or on any number of days between the minimum and the maximum.
  • a treatment course is one day or the treatment course is of a length not to exceed 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days, which are examples of a course wherein the compound selected from the table 1 ab and Table 1c is administered on two or more days over a treatment course not to exceed (i.e., no longer than) 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days, respectively.
  • the treatment course is about 2 weeks (about 14 days or 0.5 months), about 3 weeks (about 21 days), about 4 weeks (about one month), about 5 weeks, about 6 weeks (about 1.5 months), about 2 months (or about 60 days), or about 3 months (or about 90 days).
  • a treatment course is a single daily dosing of the Compound selected from the table 1 ab and Table 1c .
  • a daily dose of the compound selected from the table 1 ab and Table 1c may be as a single administration or the dose may be divided into 2, 3, 4, or 5 separate administrations to provide the total daily dose of the agent.
  • the treatment course may have one or more intervals of one or more days when the Compound selected from the table 1 ab and Table 1c , is not administered.
  • a first dose when a treatment window is between two and seven days, a first dose may be administered on the first day of the treatment window and a second dose may be administered on the third day of the course, and a third dose may be administered on the seventh day of the treatment window.
  • varying dosing schedules may be used during a particular treatment window.
  • the compound selected from the table 1 ab and Table 1c is administered daily on each consecutive day for the duration of the treatment course.
  • a daily dose may be administered as a single dose or the daily dose may be divided into 2, 3, or 4, or 5 separate administrations to provide the total daily dose of the Compound selected from the table 1 ab and Table 1c .
  • the treatment course comprises a length of time during which the compound selected from the table 1 ab and Table 1c is administered daily.
  • the compound selected from the table 1 ab and Table 1c is administered daily for 2 days.
  • the compound selected from the table 1 ab and Table 1c is administered daily for 3 days.
  • the compound selected from the table 1 ab and Table 1c is administered daily for 4 days.
  • the compound selected from the table 1 ab and Table 1c is administered daily for 5 days.
  • the compound selected from the table 1 ab and Table 1c is administered daily for 6 days.
  • the compound selected from the table 1 ab and Table 1c is administered daily for 7 days.
  • the compound selected from the table 1 ab and Table 1c is administered daily for 8 days. In still another specific embodiment, the compound selected from the table 1 ab and Table 1c is administered daily for 9 days. In yet another particular embodiment, the compound selected from the table 1 ab and Table 1c is administered daily for 10 days. In yet another particular embodiment, the compound selected from the table 1 ab and Table 1c is administered daily for 11 days. In yet another particular embodiment, the compound selected from the table 1 ab and Table 1c is administered daily for 12 days. In yet another particular embodiment, the compound selected from the table 1 ab and Table 1c is administered daily for 13 days. In yet another particular embodiment, the compound selected from the table 1 ab and Table 1c is administered daily for 14 days.
  • the treatment window (i.e., course) for each of the above examples is no longer than 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 days, respectively.
  • the compound selected from the table 1 ab and Table 1c is administered every 2 nd day (i.e., every other day) for 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days.
  • the compound selected from the table 1 ab and Table 1c is administered every 3 nd day (i.e., one day receiving the agent followed by two days without receiving the agent) for 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days.
  • the compound selected from the table 1 ab and Table 1c may be administered on every 2 nd -3 rd day during a treatment window of 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days.
  • the compound selected from the table 1 ab and Table 1c may be administered every 4 th day during a treatment course of 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days; or every 5 th day during a treatment course of 6, 7, 8, 9, 10, 11, 12, 13, or 14 days.
  • a person skilled in the art can readily appreciate the minimum numbers of days in a treatment window when the compound selected from the table 1 ab and Table 1c is administered every 6 th , 7 th , etc. day over a treatment window of a finite number of days as described herein.
  • a compound selected from the table 1 ab and Table 1c may be administered daily for a longer duration than 14 days and may be administered at least 15, 16, 17, 18, 19, 20, or at least 21 days. In other specific embodiments, the compound selected from the table 1 ab and Table 1c may be administered daily on each of the 15, 16, 17, 18, 19, 20, or 21 days. In another specific embodiment, the compound selected from the table 1 ab and Table 1c may be administered every second day during a treatment window of 15, 16, 17, 18, 19, 20, or 21 days. In another specific embodiment, the compound selected from the table 1 ab and Table 1c may be administered every third day during a treatment window of 15, 16, 17, 18, 19, 20, or 21 days.
  • the compound selected from the table 1 ab and Table 1c may be administered on every 2 nd -3 rd day during a treatment window of 15, 16, 17, 18, 19, 20, or 21 days. In yet other embodiments, the compound selected from the table 1 ab and Table 1c may be administered every 4 th day during a treatment course of 15, 16, 17, 18, 19, 20, or 21 days; or every 5 th day during a treatment course of 15, 16, 17, 18, 19, 20, or 21 days.
  • a person skilled in the art can readily appreciate the minimum numbers of days in a treatment window when the compound selected from the table 1 ab and Table 1c is administered every 6 th , 7 th , etc. day over a treatment window of a finite number of days as described herein.
  • a compound selected from the table 1 ab and Table 1c may be administered daily for a longer duration than 14 days and may be administered at least 15, 16, 17, 18, 19, 20, or at least 21 days. In other specific embodiments, the compound selected from the table 1 ab and Table 1c may be administered daily on each of the 15, 16, 17, 18, 19, 20, or 21 days. In another specific embodiment, the compound selected from the table 1 ab and Table 1c may be administered every second day during a treatment window of 15, 16, 17, 18, 19, 20, or 21 days. In another specific embodiment, the compound selected from the table 1 ab and Table 1c may be administered every third day during a treatment window of 15, 16, 17, 18, 19, 20, or 21 days.
  • the compound selected from the table 1 ab and Table 1c may be administered on every 2 nd -3 rd day during a treatment window of 15, 16, 17, 18, 19, 20, or 21 days. In yet other embodiments, the compound selected from the table 1 ab and Table 1c may be administered every 4 th day during a treatment course of 15, 16, 17, 18, 19, 20, or 21 days; or every 5 th day during a treatment course of 15, 16, 17, 18, 19, 20, or 21 days.
  • a person skilled in the art can readily appreciate the minimum numbers of days in a treatment window when the compound selected from the table 1 ab and Table 1c is administered every 6 th , 7 th , etc. day over a treatment window of a finite number of days as described herein.
  • a compound selected from the table 1 ab and Table 1c may be administered in a treatment course daily for a longer duration than 14 days or 21 days and may be administered in a treatment course of about one month, about two months, or about three months.
  • the compound selected from the table 1 ab and Table 1c may be administered daily on each of a one month, two month, or three month treatment course.
  • the compound selected from the table 1 ab and Table 1c may be administered every second day during a treatment course of about one month, about two months, or about three months.
  • the compound selected from the table 1 ab and Table 1c may be administered every third day during a treatment course of about one month, about two months, or about three months.
  • the compound selected from the table 1 ab and Table 1c may be administered on every 2 nd -3 rd day during a treatment course of about one month, about two months, or about three months.
  • the compound selected from the table 1 ab and Table 1c may be administered every 4 th day during a treatment course of about one month, about two months, or about three months; or every 5 th day during a treatment course of about one month, about two months, or about three months s.
  • a person skilled in the art can readily appreciate the minimum numbers of days in a treatment course when the compound selected from the table 1 ab and Table 1c is administered every 6 th , 7 th , etc. day over a treatment window of a finite number of days as described herein.
  • a longer treatment window with a decreased dose per day may be a treatment option for a subject.
  • the stage or severity of the senescence associated disease or disorder or other clinical factor may indicate that a longer term course may provide clinical benefit.
  • the compound selected from the table 1 ab and Table 1c is administered daily, or optionally, every other day (every 2 nd day) or every 3 rd day, or greater interval (i.e., every 4 th day, 5 th day, 6 th day) during a treatment course of about 1-2 weeks (e.g., about 5-14 days), about 1-3 weeks (e.g., about 5-21 days), about 1-4 weeks (e.g., about 5-28 days, about 5-36 days, or about 5-42 days, 7-14 days, 7-21 days, 7-28 days, 7-36 days, or 7-42 days; or 9-14 days, 9-21 days, 9-28 days, 9-36 days, or 9-42 days.
  • the treatment course is between about 1- 3 months.
  • the compound selected from the table 1 ab and Table 1c is administered daily for at least five days, and in another particular embodiment, the compound selected from the table 1 ab and Table 1c is administered daily for 5-14 days.
  • the compound selected from the table 1 ab and Table 1c is administered for at least seven days, for example, for 7-14, 7-21, 7-28 days, 7-36 days, or 7-42 days.
  • the compound selected from the table 1 ab and Table 1c is administered for at least nine days, for example, for 9-14 days, 9-21 days, 9-28 days, 9-36 days, or 9-42 days.
  • a treatment course comprising administering a compound selected from the table 1 ab and Table 1c provides clinical benefit
  • a treatment course is repeated with a time interval between each treatment course when the compound selected from the table 1 ab and Table 1c is not administered (i.e., non-treatment interval, off- drug treatment).
  • a treatment cycle as described herein and in the art comprises a treatment course followed by a non-treatment interval.
  • a treatment cycle may be repeated as often as needed. For example, a treatment cycle may be repeated at least once, at least twice, at least three times, at least four times, at least five times, or more often as needed.
  • a treatment cycle is repeated once (i.e., administration of the compound selected from the table 1 ab and Table 1c comprises 2 treatment cycles). In other certain embodiments, the treatment cycle is repeated twice or repeated 3 or more times. Accordingly, in certain embodiments, one, two, three, four, five, six, seven, eight, nine, ten, or more treatment cycles of treatment with a compound selected from the table 1 ab and Table 1c are performed.
  • a treatment course or a treatment cycle may be repeated, such as when the senescence associated disease or disorder recurs, or when symptoms or sequelae of the disease or disorder that were significantly diminished by one treatment course as described above have increased or are detectable, or when the symptoms or sequelae of the disease or disorder are exacerbated, a treatment course may be repeated.
  • a subject when the compound selected from the table 1 ab and Table 1c is administered to a subject to prevent (i.e., reduce likelihood of occurrence or development) or to delay onset, progression, or severity of senescence associated disease or disorder, a subject may receive the compound selected from the table 1 ab and Table 1c over two or more treatment cycles.
  • one cycle of treatment is followed by a subsequent cycle of treatment.
  • Each treatment course of a treatment cycle or each treatment course of two or more treatment cycles are typically the same in duration and dosing of the Compound selected from the table 1 ab and Table 1c .
  • the duration and dosing of the compound selected from the table 1 ab and Table 1c during each treatment course of a treatment cycle may be adjusted as determined by a person skilled in the medical art depending, for example, on the particular disease or disorder being treated, the compound selected from the table 1 ab and Table 1c being administered, the health status of the patient and other relevant factors, which are discussed in greater detail herein.
  • each treatment course of two or more treatment cycles are independent and the same or different; and each non-treatment interval of each treatment cycle is independent and the same or different.
  • each course of treatment in a treatment cycle is separated by a time interval of days, weeks, or months without treatment with a compound selected from the table 1 ab and Table 1c (i.e., non-treatment time interval or off-drug interval; called non-treatment interval herein).
  • the non-treatment interval (such as days, weeks, months) between one treatment course and a subsequent treatment course is typically greater than the longest time interval (i.e., number of days) between any two days of administration in the treatment course.
  • the non-treatment interval between two treatment courses is greater than 2 days, such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days or about 3 weeks, about 4 weeks, about 6 weeks, or about 2 months or longer as described herein.
  • the non-treatment interval between two treatment courses is about 5 days, about 1 week, about 2 weeks, about 3 weeks, about 1 month, about 6 weeks, about 2 months (8 weeks), about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 12 months (about 1 year), about 18 months (about 1.5 years), or longer.
  • the non-treatment interval is about 2 years or about 3 years.
  • the non-treatment time interval is at least about 14 days, at least about 21 days, at least about 1 month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, or at least about 1 year.
  • a course of treatment (whether daily, every other day, every 3 rd day, or other interval between administrations within the treatment course as described above (e.g., 1-14 days, 2-14 days, 2-21 days, or 1- 21 days)) is administered about every 14 days (i.e., about every 2 weeks) (i.e., 14 days without compound selected from the table 1 ab and Table 1c treatment), about every 21 days (i.e., about every 3 weeks), about every 28 days (i.e., about every 4 weeks), about every one month, about every 36 days, about every 42 days, about every 54 days, about every 60 days, or about every month (about every 30 days), about every two months (about every 60 days), about every quarter (about every 90 days), or about semi-annually (about every 180 days).
  • a course of treatments (e.g., byway of non-limiting example, administration on at least one day or on at least two days during a course for about 2-21 days, about 2-14, days, about 5-14 days, about 7-14 days, about 9-14 days, about 5-21 days, about 7-21 days, about 9-21 days) is administered every 28 days, every 36 days, every 42 days, every 54 days, every 60 days, or every month (about every 30 days), every two months (about every 60 days), every quarter (about every 90 days), or semi-annually (about every 180 days), or about every year (about 12 months).
  • a course of treatment (such as by way of non-limiting examples, e.g., for about 5-28 days, about 7-28 days, or about 9-28 days whether daily, every other day, every 3 rd day, or other interval between administrations within the treatment course) is administered every 36 days, 42 days, 54 days, 60 days, or every month (about every 30 days), every two months (about every 60 days), every quarter (about every 90 days), or semi-annually (about every 180 days).
  • a course of treatment (e.g., for about 5-36 days, 7-36 days, or 9-36 days whether daily, every other day, every 3 rd day, or other interval between administrations within the treatment course) is administered every 42 days, 54 days, 60 days, or every month (about every 30 days), every two months (about every 60 days), every quarter (about every 90 days), or semi-annually (about every 180 days), or about every year (about 12 months).
  • the treatment course is one day and the non-treatment interval is at least about 14 days, about 21 days, about 1 month, about 2 months (8 weeks), about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 12 months (about 1 year), about 18 months (about 1.5 years), or longer.
  • the treatment course is at least two days or is at least 3 days and no longer than 10 days
  • the non-treatment interval is at least about 14 days, about 21 days, about 1 month, about 2 months (8 weeks), about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 12 months (about 1 year), about 18 months (about 1.5 years), or longer.
  • the treatment course is at least three days and no longer than 10 days, no longer than 14 days, or no longer than 21 days
  • the non-treatment interval is at least about 14 days, about 21 days, about 1 month, about 2 months (8 weeks), about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 12 months (about 1 year), about 18 months (about 1.5 years), or longer.
  • a treatment course (e.g., for about 5-42, 7-42, or 9-42 days whether daily, every other day, every 3 rd day, or other interval between administrations within the treatment course) is administered every 42 days, 60 days, or every month (about every 30 days), every two months (about every 60 days), every quarter (about every 90 days), or semi-annually (about every 180 days), or about every year (about 12 months).
  • the compound selected from the table 1 ab and Table 1c is administered daily for 5-14 days every 14 days (about every 2 weeks), or every 21-42 days.
  • the compound selected from the table 1 ab and Table 1c is administered daily for 5-14 days quarterly.
  • the compound selected from the table 1 ab and Table 1c is administered daily for 7-14 days every 21-42 days. In another particular embodiment, the compound selected from the table 1 ab and Table 1c is administered daily for 7-14 days quarterly. In still other particular embodiments, the compound selected from the table 1 ab and Table 1c is administered daily for 9-14 days every 21-42 days or every 9-14 days quarterly. In still other embodiments, the non-treatment interval may vary between treatment courses. By way of non-limiting example, the non-treatment interval may be 14 days after the first course of treatment and may be 21 days or longer after the second, third, or fourth (or more) course of treatment. In other particular embodiments, the compound selected from the table 1 ab and Table 1c is administered to the subject in need thereof once every 0.5-12 months.
  • the compound selected from the table 1 ab and Table 1c is administered to the subject in need once every 4-12 months.
  • a compound selected from the table 1 ab and Table 1c is administered to a subject to reduce the likelihood or the risk that the subject will develop a particular disorder or to delay onset of one or more symptoms of a senescence-associated disease or disorder.
  • the compound selected from the table 1 ab and Table 1c is administered for one or more days (e.g., any number of consecutives days between and including 2-3, -4, -5, -6, -7, -8, -9, -10, -11, -12, -13, -14, -15, -16, -17, -18, -19, -20, and 2-21 days) every 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months.
  • the compound selected from the table 1 ab and Table 1c is administered for one or more days (e.g., any number of consecutives days between and including 1-9 days) every 5 or 6 months.
  • periodic administration of the compound selected from the table 1 ab and Table 1c kills newly formed senescent cells and thereby reduces (decreases, diminishes) the total number of senescent cells accumulating in the subject.
  • the total number of senescent cells accumulating in the subject is decreased or inhibited by administering the compound selected from the table 1 ab and Table 1c once or twice weekly or according to any of the other treatment courses described above.
  • the total daily dose of a compound selected from the table 1 ab and Table 1c may be delivered as a single dose or as multiple doses on each day of administration.
  • the dose of a compound selected from the table 1 ab and Table 1c administered on a single day may be less than the daily dose administered if only a single treatment course is intended to be administered.
  • method for treating a senescence-associated disease or disorder comprising administering to a subject in need thereof a small molecule compound selected from the table 1 ab and Table 1c that selectively kills senescent cells; wherein the senescence-associated disease or disorder is not a cancer, and wherein the compound selected from the table 1 ab and Table 1c is administered within one or two treatment cycles, typically two treatment cycles.
  • the non-treatment interval is at least 2 weeks and each treatment course is no longer than 3 months.
  • methods for selectively killing a senescent cell comprising contacting the senescent cell with a compound selected from the table 1 ab and Table 1c described herein (i.e., facilitating interaction or in some manner allowing the senescent cell and compound selected from the table 1 ab and Table 1c to interact) under conditions and for a time sufficient to kill the senescent cell.
  • the agent selectively kills senescent cells over non-senescent cells (i.e., the agent selectively kills senescent cells compared with killing of non-senescent cells).
  • the senescent cell to be killed is present in a subject (e.g., a human or non-human animal).
  • the Compound selected from the table 1 ab and Table 1c (s) may be administered to the subject according to the treatment cycles, treatment courses, and non-treatment intervals described above and herein.
  • a single (i.e., only, sole) compound selected from the table 1 ab and Table 1c is administered to the subject for treating a senescence-associated disease or disorder.
  • administration of a single compound selected from the table 1 ab and Table 1c may be sufficient and clinically beneficial to treat a senescence-associated disease or disorder.
  • a compound selected from the table 1 ab and Table 1c is administered as a monotherapy and is the single (i.e., only, sole) active agent administered to the subject for treating the condition or disease.
  • Medications that are not necessarily excluded from administration to the subject when a compound selected from the table 1 ab and Table 1c is administered as a monotherapy include, by way of non-limiting examples, medications for other purposes such as palliative care or comfort (e.g., aspirin, acetominophen, ibuprofen, or prescription pain-killers; anti-itching topical medications) or for treating a different disease or condition, especially if the other medications are not senolytic agents, such as drugs for lowering cholesterol, statins, eye wetting agents, and other such medications familiar to a person skilled in the medical art.
  • the methods for treating a senescence associated disease or disorder comprise administering the compound selected from the table 1 ab and Table 1c in one or two or more treatment cycles, and the total dose of the compound selected from the table 1 ab and Table 1c administered during each treatment course, each treatment cycle, and/or cumulatively over two or more treatment cycles is an amount less than the amount effective for a cancer treatment.
  • the amount of such a compound selected from the table 1 ab and Table 1c administered to a subject over a given time period (such as one week, two weeks, one month, six months, one year) for treating a senescence associated disease or disorder, for example, may be about from a 20-fold decrease to about a 5000-fold decrease in total amount compared with the total amount of the same agent administered to a subject who is receiving the agent for treatment of a cancer.
  • the fold decrease in the amount (i.e., lesser amount) of the compound selected from the table 1 ab and Table 1c administered over a given time period (i.e., number of days, months, years) for treating a senescence associated disease or disorder may be about a 20-fold decrease, about a 25-fold decrease, about a 30-fold decrease, about a 40-fold decrease, about a 50-fold decrease, about a 60-fold decrease, about a 75-fold decrease, about a 100-fold decrease, about a 125-fold decrease, about a 150-fold decrease, about a 175-fold decrease, about a 200-fold decrease, about a 300-fold decrease, about a 400-fold decrease, about a 500-fold decrease, about a 750-fold decrease, about a 1000-fold decrease, about a 1250-fold decrease, about a 1500-fold decrease, about a 1750-fold decrease, about a 2000-fold decrease, about a 2250- fold decrease, about a 2500-fold decrease, about
  • a lower dose required for treating a senescence associated disease may also be attributable to the route of administration.
  • a compound selected from the table 1 ab and Table 1c when used for treating a senescence-associated pulmonary disease or disorder (e.g., COPD, IPF), the compound selected from the table 1 ab and Table 1c may be delivered directly to the lungs (e.g., by inhalation, by intubation, intranasally, or intratracheally), and a lower dose per day and/or per treatment course is required than if the agent were administered orally.
  • the compound selected from the table 1 ab and Table 1c when a compound selected from the table 1 ab and Table 1c is used for treating osteoarthritis or a senescence- associated dermatological disease or disorder, the compound selected from the table 1 ab and Table 1c may be delivered directly to the osteoarthritic joint (e.g., intra-articularly, intradermally, topically, transdermally) or to the skin (e.g., topically, subcutaneously, intradermally, transdermally), respectively, at a lower does per day and/or per treatment course than if the compound selected from the table 1 ab and Table 1c were administered orally.
  • the osteoarthritic joint e.g., intra-articularly, intradermally, topically, transdermally
  • the skin e.g., topically, subcutaneously, intradermally, transdermally
  • the dose of the compound selected from the table 1 ab and Table 1c per day may be the same amount as administered to a patient for treating a cancer; however, the amount of the agent that is delivered over a treatment course or treatment cycle is significantly less than the amount administered to a subject who receives the appropriate amount of the agent for treating a cancer.
  • the methods described herein comprise using the compound selected from the table 1 ab and Table 1c in an amount that is a reduced amount compared with the amount that may be delivered systemically, for example, orally or intravenously to a subject who receives the compound selected from the table 1 ab and Table 1c when the agent is used for treating a cancer.
  • methods of treating a senescence-associated disease or disorder by selectively killing senescent cells comprises administering the compound selected from the table 1 ab and Table 1c at a dose that is at least 10% (i.e., one-tenth), at least 20% (one-fifth), 25% (one-fourth), 30%-33% (about one-third), 40% (two-fifths), or at least 50% (half) of the dose that is administered to a subject who has cancer for killing cancer cells during a treatment course, a treatment cycle, or two or more treatment cycles that form the cancer therapy protocol (i.e., regimen).
  • a dose that is at least 10% (i.e., one-tenth), at least 20% (one-fifth), 25% (one-fourth), 30%-33% (about one-third), 40% (two-fifths), or at least 50% (half) of the dose that is administered to a subject who has cancer for killing cancer cells during a treatment course, a treatment cycle, or two or more treatment cycles that form the cancer therapy protocol (i.
  • the dose of the Compound selected from the table 1 ab and Table 1c (s) used in the methods described herein is at least 60%, 70%, 80%, 85%, 90%, or 95% of the dose that is administered to a subject who has cancer.
  • the therapeutic regimen, comprising the dose of compound selected from the table 1 ab and Table 1c and schedule and manner of administration that may be used for treating a senescence-associated disorder or disease is also a regimen insufficient to be significantly cytotoxic to non-senescent cells.
  • the dose of the Compound selected from the table 1 ab and Table 1c used in the methods described herein is at least 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 115%, 115%, 130%, 150%, 175%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, or 1000% of the lowest dose that is administered to a subject in Phase 1 clinical trial and in formulation, root of administration and regimen as it were in such Phase 1 clinical trial.
  • the dose of the Compound selected from the table 1 ab and Table 1c used in the methods described herein is at least 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 115%, 115%, 130%, 150%, 175%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, or 1000% of the highest dose that is administered to a subject in Phase 2 clinical trial and in formulation, root of administration and regimen as it were in such Phase 2 clinical trial for any one of the others indications [0063] In other particular embodiments, the dose of the Compound selected from the table 1 ab and Table 1c used in the methods described herein is at least 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 115%, 115%, 130%, 150%, 17
  • the dose of the Compound selected from the table 1 ab and Table 1c used in the methods described herein is at least 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 115%, 115%, 130%, 150%, 175%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, or 1000% of the highest dose that is approved and in formulation, root of administration and regimen as it were approved for other any one of others indications.
  • a method for treating a senescence-associated disease or disorder that is not a cancer comprises administering to a subject in need thereof a therapeutically effective amount of a small molecule compound selected from the table 1 ab and Table 1c that selectively kills senescent cells (i.e., selectively kills senescent cells over non-senescent cells or compared with non-senescent cells) and which agent is cytotoxic to cancer cells, wherein the compound selected from the table 1 ab and Table 1c is administered within at least one treatment cycle, which treatment cycle comprises a treatment course followed by a non-treatment interval.
  • the total dose of the compound selected from the table 1 ab and Table 1c administered during the treatment course, and/or the total dose of the compound selected from the table 1 ab and Table 1c administered during the treatment cycle, and/or the total dose of the compound selected from the table 1 ab and Table 1c administered during two or more treatment cycles is an amount less than the amount effective for a cancer treatment.
  • the compound selected from the table 1 ab and Table 1c is administered as a monotherapy, and is the single active compound selected from the table 1 ab and Table 1c administered to the subject for treating the disease or disorder.
  • the number of days in the treatment course and the treatment interval are described in detail herein.
  • a method for treating a senescence-associated disease or disorder, wherein the senescence-associated disease is not cancer and the method comprises administering to a subject in need thereof a compound selected from the table 1 ab and Table 1c or small molecule senolytic compound that selectively kills senescent cells, and the administration is for a short duration (e.g., shorter than may be used for a particular agent for treating a cancer), such as a single day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, or 15 days.
  • a short duration e.g., shorter than may be used for a particular agent for treating a cancer
  • this treatment course on any number of days between 1-15 days is a single treatment course and is not repeated.
  • a compound selected from the table 1 ab and Table 1c is administered for 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days, or 31 days as a single treatment course that is not repeated.
  • a senescence-associated disease or disorder may also be called herein a senescent cell- associated disease or disorder.
  • Senescence-associated diseases and disorders include, for example, cardiovascular diseases and disorders, inflammatory diseases and disorders, autoimmune diseases and disorders, pulmonary diseases and disorders, eye diseases and disorders, metabolic diseases and disorders, neurological diseases and disorders (e.g., neurodegenerative diseases and disorders); age-related diseases and disorders induced by senescence; skin conditions; age-related diseases; dermatological diseases and disorders; and transplant related diseases and disorders.
  • a prominent feature of aging is a gradual loss of function, or degeneration that occurs at the molecular, cellular, tissue, and organismal levels.
  • Age-related degeneration gives rise to well-recognized pathologies, such as sarcopenia, atherosclerosis and heart failure, osteoporosis, pulmonary insufficiency, renal failure, neurodegeneration (including macular degeneration, Alzheimer's disease, and Parkinson's disease), and many others.
  • pathologies such as sarcopenia, atherosclerosis and heart failure, osteoporosis, pulmonary insufficiency, renal failure, neurodegeneration (including macular degeneration, Alzheimer's disease, and Parkinson's disease), and many others.
  • pathologies such as sarcopenia, atherosclerosis and heart failure, osteoporosis, pulmonary insufficiency, renal failure, neurodegeneration (including macular degeneration, Alzheimer's disease, and Parkinson's disease), and many others.
  • age-related pathologies generally rise with approximately exponential kinetics beginning at about the mid-point of the species-specific life span (e.g., 50-60 years of age for humans).
  • Examples of senescence-associated conditions, disorders, or diseases that may be treated by administering any one of the Compounds selected from the table 1 ab and Table 1c described herein according to the methods described herein include, cognitive diseases (e.g., mild cognitive impairment (MCI), Alzheimer's disease and other dementias; Huntington's disease); cardiovascular disease (e.g., atherosclerosis, cardiac diastolic dysfunction, aortic aneurysm, angina, arrhythmia, cardiomyopathy, congestive heart failure, coronary artery disease, myocardial infarction, endocarditis, hypertension, carotid artery disease, peripheral vascular diseases, cardiac stress resistance, cardiac fibrosis); metabolic diseases and disorders (e.g., obesity, diabetes, metabolic syndrome); motor function diseases and disorders (e.g., Parkinson's disease, motor neuron dysfunction (MND); Huntington'
  • any one or more of the diseases or disorders described above or herein may be excluded.
  • methods are provided for treating a senescence-associated disease or disorder by killing senescent cells (i.e., established senescent cells) associated with the disease or disorder in a subject who has the disease or disorder by administering a Compound selected from the table 1 ab and Table 1c , wherein the disease or disorder is osteoarthritis; idiopathic pulmonary fibrosis; chronic obstructive pulmonary disease (COPD); or atherosclerosis.
  • senescent cells i.e., established senescent cells
  • COPD chronic obstructive pulmonary disease
  • Subjects i.e., patients, individuals (human or non-human animals) who may benefit from use of the methods described herein that comprise administering a compound selected from the table 1 ab and Table 1c include those who may also have a cancer.
  • the subject treated by these methods may be considered to be in partial or complete remission (also called cancer remission).
  • the Compounds selected from the table 1 ab and Table 1c for use in methods for selective killing of senescent cells are not intended to be used as a treatment for cancer, that is, in a manner that kills or destroys the cancer cells in a statistically significant manner.
  • the methods disclosed herein do not encompass use of the Compounds selected from the table 1 ab and Table 1c in a manner that would be considered a primary therapy for the treatment of a cancer. Even though a Compound selected from the table 1 ab and Table 1c , alone or with other chemotherapeutic or radiotherapy agents, are not used in a manner that is sufficient to be considered as a primary cancer therapy, the methods and Compounds selected from the table 1 ab and Table 1c described herein may be used in a manner (e.g., a short term course of therapy) that is useful for inhibiting metastases.
  • the subject to be treated with the compound selected from the table 1 ab and Table 1c does not have a cancer (i.e., the subject has not been diagnosed as having a cancer by a person skilled in the medical art).
  • Cardiovascular Diseases and Disorders the senescence-associated disease or disorder treated by the methods, comprising administering a compound selected from the table 1 ab and Table 1c , described herein is a cardiovascular disease.
  • the cardiovascular disease may be any one or more of angina, arrhythmia, atherosclerosis, cardiomyopathy, congestive heart failure, coronary artery disease (CAD), carotid artery disease, endocarditis, heart attack (coronary thrombosis, myocardial infarction [MI]), high blood pressure/hypertension, aortic aneurysm, brain aneurysm, cardiac fibrosis, cardiac diastolic dysfunction, hypercholesterolemia/hyperlipidemia, mitral valve prolapse, peripheral vascular disease (e.g., peripheral artery disease (PAD)), cardiac stress resistance, and stroke.
  • CAD coronary artery disease
  • MI myocardial infarction
  • methods are provided for treating senescence-associated cardiovascular disease that is associated with or caused by arteriosclerosis (i.e., hardening of the arteries).
  • such methods comprise administering a compound selected from the table 1 ab and Table 1c .
  • the cardiovascular disease may be any one or more of atherosclerosis (e.g., coronary artery disease (CAD) and carotid artery disease); angina, congestive heart failure, and peripheral vascular disease (e.g., peripheral artery disease (PAD)), wherein in some embodiments such methods comprise administering a compound selected from the table 1 ab and Table 1c .
  • atherosclerosis e.g., coronary artery disease (CAD) and carotid artery disease
  • angina CAD
  • congestive heart failure e.g., congestive heart failure
  • peripheral vascular disease e.g., peripheral artery disease (PAD)
  • the methods for treating a cardiovascular disease that is associated with or caused by arteriosclerosis may reduce the likelihood of occurrence of high blood pressure/hypertension, angina, stroke, and heart attack (i.e., coronary thrombosis, myocardial infarction (MI)).
  • methods for stabilizing atherosclerotic plaque(s) in a blood vessel (e.g., artery) of a subject thereby reducing the likelihood of occurrence or delaying the occurrence of a thrombotic event, such as stroke or MI.
  • these methods comprising administration of a compound selected from the table 1 ab and Table 1c reduce (i.e., cause decrease of) the lipid content of an atherosclerotic plaque in a blood vessel (e.g., artery) of the subject and/or increase the fibrous cap thickness (i.e., cause an increase, enhance or promote thickening of the fibrous cap).
  • methods are provided for inhibiting the formation of atherosclerotic plaques (or reducing, diminishing, causing decrease in formation of atherosclerotic plaques) by administering a Compound selected from the table 1 ab and Table 1c .
  • methods are provided for reducing (decreasing, diminishing) the amount (i.e., level) of plaque.
  • Reduction in the amount of plaque in a blood vessel (e.g., artery) may be determined, for example, by a decrease in surface area of the plaque, or by a decrease in the extent or degree (e.g., percent) of occlusion of a blood vessel (e.g., artery), which can be determined by angiography or other visualizing methods used in the cardiovascular art.
  • this invention suggested a method of atheromatous plaque stabilization comprising administering to the subject any one of the Compounds selected from the table 1 ab and Table 1c .
  • Subjects suffering from cardiovascular disease can be identified using standard diagnostic methods known in the art for cardiovascular disease.
  • Atherosclerosis and other cardiovascular disease is based on symptoms (e.g., chest pain or pressure (angina), numbness or weakness in arms or legs, difficulty speaking or slurred speech, drooping muscles in face, leg pain, high blood pressure, kidney failure and/or erectile dysfunction), medical history, and/or physical examination of a patient. Diagnosis may be confirmed by angiography, ultrasonography, or other imaging tests.
  • Subjects at risk of developing cardiovascular disease include those having any one or more of predisposing factors, such as a family history of cardiovascular disease and those having other risk factors (i.e., predisposing factors) such as high blood pressure, dyslipidemia, high cholesterol, diabetes, obesity and cigarette smoking, sedentary lifestyle, and hypertension.
  • the cardiovascular disease that is a senescence cell associated disease/disorder is atherosclerosis.
  • the effectiveness of one or more Compounds selected from the table 1 ab and Table 1c for treating or preventing (i.e., reducing or decreasing the likelihood of developing or occurrence of) a cardiovascular disease (e.g., atherosclerosis) can readily be determined by a person skilled in the medical and clinical arts.
  • One or any combination of diagnostic methods including physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods described herein and practiced in the art (e.g., angiography, electrocardiography, stress test, non-stress test), may be used for monitoring the health status of the subject.
  • the effects of the treatment of a compound selected from the table 1 ab and Table 1c or pharmaceutical composition comprising same can be analyzed using techniques known in the art, such as comparing symptoms of patients suffering from or at risk of cardiovascular disease that have received the treatment with those of patients without such a treatment or with placebo treatment.
  • a senescence-associated disease or disorder is an inflammatory disease or disorder, such as by way of non-limiting example, osteoarthritis, that may be treated or prevented (i.e., likelihood of occurrence is reduced) according to the methods described herein that comprise administration of a Compound selected from the table 1 ab and Table 1c .
  • inflammatory or autoimmune diseases or disorders that may be treated by administering a compound selected from the table 1 ab and Table 1c such as the inhibitors and antagonists described herein include osteoporosis, psoriasis, oral mucositis, rheumatoid arthritis, inflammatory bowel disease, eczema, kyphosis, herniated intervertebral disc, and the pulmonary diseases, COPD and idiopathic pulmonary fibrosis.
  • a senescence-associated disease or disorder is age- and high fat- diet-induced vascular calcification and hyporeactivity, and radiation-induced muscle dysfunction.
  • a compound selected from the table 1 ab and Table 1c prevents (i.e., reduces the likelihood of occurrence), reduces or inhibits loss or erosion of proteoglycan layers in a joint, reduces inflammation in the affected joint, and promotes (i.e., stimulates, enhances, induces) production of collagen (e.g., type 2 collagen).
  • administration of a compound selected from the table 1 ab and Table 1c causes a reduction in the amount (i.e., level) of inflammatory cytokines, such as IL-6, produced in a joint and inflammation is reduced.
  • Methods are provided herein for treating osteoarthritis, and/or for selectively killing senescent cells in an osteoarthritic joint of a subject, and/or inducing collagen (such as Type 2 collagen) production in the joint of a subject in need thereof by administering at least one compound selected from the table 1 ab and Table 1c (which may be combined with at least one pharmaceutically acceptable excipient to form a pharmaceutical composition) to the subject.
  • a compound selected from the table 1 ab and Table 1c also may be used for decreasing (inhibiting, reducing) production of metalloproteinase 13 (MMP-13), which degrades collagen in a joint, and for restoring proteoglycan layer or inhibiting loss and/or degradation of the proteoglycan layer.
  • Treatment with the compound selected from the table 1 ab and Table 1c thereby also prevents (i.e., reduces likelihood of occurrence of), inhibits, or decreases erosion, or slows (i.e., decreases rate) erosion of the bone.
  • the compound selected from the table 1 ab and Table 1c is administered directly to an osteoarthritic joint (e.g., by intra-articularly, topical, transdermal, intradermal, or subcutaneous delivery). Treatment with a compound selected from the table 1 ab and Table 1c can also restore, improve, or inhibit deterioration of strength of a joint.
  • the methods comprising administering a compound selected from the table 1 ab and Table 1c can reduce joint pain and are therefore useful for pain management of osteoarthritic joints.
  • diagnostic methods including physical examination (such as determining tenderness, swelling or redness of the affected joint), assessment and monitoring of clinical symptoms (such as pain, stiffness, mobility), and performance of analytical tests and methods described herein and practiced in the art (e.g., determining the level of inflammatory cytokines or chemokines; X-ray images to determine loss of cartilage as shown by a narrowing of space between the bones in a joint; magnetic resonance imaging (MRI), providing detailed images of bone and soft tissues, including cartilage), may be used for monitoring the health status of the subject.
  • physical examination such as determining tenderness, swelling or redness of the affected joint
  • clinical symptoms such as pain, stiffness, mobility
  • analytical tests and methods described herein and practiced in the art e.g., determining the level of inflammatory cytokines or chemokines; X-ray images to determine loss of cartilage as shown by a narrowing of space between the bones in a joint; magnetic resonance imaging (MRI), providing detailed images of bone and soft tissues, including cartilage
  • Compounds selected from the table 1 ab and Table 1c can be analyzed by comparing symptoms of patients suffering from or at risk of an inflammatory disease or disorder, such as osteoarthritis, who have received the treatment with those of patients who have not received such a treatment or who have received a placebo treatment.
  • Compounds selected from the table 1 ab and Table 1c may be used for treating and/or preventing (i.e., decreasing or reducing the likelihood of occurrence) rheumatoid arthritis (RA).
  • Dysregulation of innate and adaptive immune responses characterize rheumatoid arthritis (RA), which is an autoimmune disease the incidence of which increases with age.
  • Rheumatoid arthritis is a chronic inflammatory disorder that typically affects the small joints in hands and feet. Whereas osteoarthritis results from, at least in part, wear and tear of a joint, rheumatoid arthritis affects the lining of joints, resulting in a painful swelling that can lead to bone erosion and joint deformity. RA can sometimes also affect other organs of the body, such as the skin, eyes, lungs and blood vessels. RA can occur in a subject at any age; however, RA usually begins to develop after age 40. The disorder is much more common in women. In certain embodiments of the methods described herein, RA is excluded.
  • Chronic inflammation may also contribute to other age-related or aging related diseases and disorders, such as kyphosis and osteoporosis.
  • Kyphosis is a severe curvature in the spinal column, and it is frequently seen with normal and premature aging. Age-related kyphosis often occurs after osteoporosis weakens spinal bones to the point that they crack and compress. A few types of kyphosis target infants or teens. Severe kyphosis can affect lungs, nerves, and other tissues and organs, causing pain and other problems. Kyphosis has been associated with cellular senescence.
  • Characterizing the capability of a compound selected from the table 1 ab and Table 1c for treating kyphosis may be determined in pre-clinical animal models used in the art.
  • TTD mice develop kyphosis (see, e.g., de Boer et al. (2002) Science 296: 1276-1279); other mice that may be used include BubR1 H/H mice, which are also known to develop kyphosis (see, e.g., Baker et al. (2011) Nature 479: 232-36). Kyphosis formation is visually measured over time.
  • the level of senescent cells decreased by treatment with the compound selected from the table 1 ab and Table 1c can be determined by detecting the presence of one or more senescent cell associated markers such as by SA- ⁇ -GAL staining.
  • Osteoporosis is a progressive bone disease that is characterized by a decrease in bone mass and density that may lead to an increased risk of fracture. Bone mineral density (BMD) is reduced, bone microarchitecture deteriorates, and the amount and variety of proteins in bone are altered. Osteoporosis is typically diagnosed and monitored by a bone mineral density test. Post-menopausal women or women who have reduced estrogen are most at risk. While both men and women over 75 are at risk, women are twice as likely to develop osteoporosis than men.
  • an inflammatory/autoimmune disorder that may be treated or prevented (i.e., likelihood of occurrence is reduced) with the Compounds selected from the table 1 ab and Table 1c described herein includes irritable bowel syndrome (IBS) and inflammatory bowel diseases, such as ulcerative colitis and Crohn's disease.
  • IBS irritable bowel syndrome
  • IBD Inflammatory bowel disease
  • IBD involves chronic inflammation of all or part of the digestive tract. In addition to life-threatening complications arising from IBD, the disease can be painful and debilitating.
  • Ulcerative colitis is an inflammatory bowel disease that causes long-lasting inflammation in part of the digestive tract. Symptoms usually develop over time, rather than suddenly. Ulcerative colitis usually affects only the innermost lining of the large intestine (colon) and rectum. Crohn's disease is an inflammatory bowel disease that causes inflammation anywhere along the lining of your digestive tract, and often extends deep into affected tissues. This can lead to abdominal pain, severe diarrhea, and malnutrition. The inflammation caused by Crohn's disease can involve different areas of the digestive tract.
  • Diagnosis and monitoring of the diseases is performed according to methods and diagnostic tests routinely practiced in the art, including blood tests, colonoscopy, flexible sigmoidoscopy, barium enema, CT scan, MRI, endoscopy, and small intestine imaging.
  • the methods described herein may be useful for treating a subject who has herniated intervertebral discs.
  • such methods comprise administering Compounds selected from the table 1 ab and Table 1c .
  • Subjects with these herniated discs exhibit elevated presence of cell senescence in the blood and in vessel walls. Symptoms of a herniated intervertebral disc may include pain, numbness or tingling, or weakness in an arm or leg.
  • inflammatory or autoimmune diseases that may be treated or prevented (i.e., likelihood of occurrence is reduced) by using a compound selected from the table 1 ab and Table 1c include eczema, psoriasis, osteoporosis, and pulmonary diseases (e.g., chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), asthma), inflammatory bowel disease, and mucositis (including oral mucositis, which in some instances is induced by radiation).
  • COPD chronic obstructive pulmonary disease
  • IPF idiopathic pulmonary fibrosis
  • asthma inflammatory bowel disease
  • mucositis including oral mucositis, which in some instances is induced by radiation
  • the senescent cell associated disorder is an inflammatory disorder of the skin, such as by way of a non-limiting examples, psoriasis and eczema that may be treated or prevented (i.e., likelihood of occurrence is reduced) according to the methods described herein that comprise administration of a Compound selected from the table 1 ab and Table 1c .
  • Psoriasis is characterized by abnormally excessive and rapid growth of the epidermal layer of the skin.
  • a diagnosis of psoriasis is usually based on the appearance of the skin. Skin characteristics typical for psoriasis are scaly red plaques, papules, or patches of skin that may be painful and itch.
  • skin characteristics typical for psoriasis are scaly red plaques, papules, or patches of skin that may be painful and itch.
  • IL-6 a key component of the SASP.
  • Eczema is an inflammation of the skin that is characterized by redness, skin swelling, itching and dryness, crusting, flaking, blistering, cracking, oozing, or bleeding.
  • Compounds selected from the table 1 ab and Table 1c for treatment of psoriasis and eczema and monitoring of a subject who receives such a compound selected from the table 1 ab and Table 1c can be readily determined by a person skilled in the medical or clinical arts.
  • diagnostic methods including physical examination (such as skin appearance), assessment of monitoring of clinical symptoms (such as itching, swelling, and pain), and performance of analytical tests and methods described herein and practiced in the art (i.e., determining the level of pro-inflammatory cytokines).
  • Compound selected from the table 1 ab and Table 1c is effective in alleviating of at least one symptoms of at least one inflammatory disorder.
  • Other immune disorders or conditions that may be treated or prevented (i.e., likelihood of occurrence is reduced) with a compound selected from the table 1 ab and Table 1c include conditions resulting from a host immune response to an organ transplant (e.g., kidney, bone marrow, liver, lung, or heart transplant), such as rejection of the transplanted organ.
  • organ transplant e.g., kidney, bone marrow, liver, lung, or heart transplant
  • the compound selected from the table 1 ab and Table 1c may be used for treating or reducing the likelihood of occurrence of graft-vs-host disease.
  • methods are provided for treating ore preventing (i.e., reducing the likelihood of occurrence of) a senescence-associated disease or disorder that is a pulmonary disease or disorder by killing senescent cells (i.e., established senescent cells) associated with the disease or disorder in a subject who has the disease or disorder by administering a Compound selected from the table 1 ab and Table 1c .
  • Senescence associated pulmonary diseases and disorders include, for example, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), asthma, cystic fibrosis, bronchiectasis, and emphysema.
  • such methods comprise administration of a Compound selected from the table 1 ab and Table 1c .
  • COPD is a lung disease defined by persistently poor airflow resulting from the breakdown of lung tissue (emphysema) and the dysfunction of the small airways (obstructive bronchiolitis).
  • Primary symptoms of COPD include shortness of breath, wheezing, chest tightness, chronic cough, and excess sputum production.
  • Elastase from cigarette smoke-activated neutrophils and macrophages disintegrates the extracellular matrix of alveolar structures, resulting in enlarged air spaces and loss of respiratory capacity.
  • COPD is most commonly caused by tobacco smoke (including cigarette smoke, cigar smoke, secondhand smoke, pipe smoke), occupational exposure (e.g., exposure to dust, smoke or fumes), and pollution, occurring over decades thereby implicating aging as a risk factor for developing COPD.
  • Compound selected from the table 1 ab and Table 1c is effective in alleviating of at least one symptoms of COPD.
  • Pulmonary fibrosis is a chronic and progressive lung disease characterized by stiffening and scarring of the lung, which may lead to respiratory failure, lung cancer, and heart failure. Fibrosis is associated with repair of epithelium. Fibroblasts are activated, production of extracellular matrix proteins is increased, and transdifferentiation to contractile myofibroblasts contribute to wound contraction.
  • a provisional matrix plugs the injured epithelium and provides a scaffold for epithelial cell migration, involving an epithelial-mesenchymal transition (EMT).
  • EMT epithelial-mesenchymal transition
  • Blood loss associated with epithelial injury induces platelet activation, production of growth factors, and an acute inflammatory response.
  • the epithelial barrier heals and the inflammatory response resolves.
  • the fibroblast response continues, resulting in unresolved wound healing. Formation of fibroblastic foci is a feature of the disease, reflecting locations of ongoing fibrogenesis. As the name connotes, the etiology of IPF is unknown.
  • Subjects at risk of developing pulmonary fibrosis include those exposed to environmental or occupational pollutants, such as asbestosis and silicosis; who smoke cigarettes; having some typical connective tissue diseases such as rheumatoid arthritis, SLE and scleroderma; having other diseases that involve connective tissue, such as sarcoidosis and Wegener's granulomatosis; having infections; taking certain medications (e.g., amiodarone, bleomycin, busufan, methotrexate, and nitrofurantoin); those subject to radiation therapy to the chest; and those whose family member has pulmonary fibrosis.
  • environmental or occupational pollutants such as asbestosis and silicosis
  • who smoke cigarettes having some typical connective tissue diseases such as rheumatoid arthritis, SLE and scleroderma; having other diseases that involve connective tissue, such as sarcoidosis and Wegener's granulomatosis; having infections; taking certain medications (e.g., amiodarone
  • Symptoms of COPD may include any one of shortness of breath, especially during physical activities; wheezing; chest tightness; having to clear your throat first thing in the morning because of excess mucus in the lungs; a chronic cough that produces sputum that may be clear, white, yellow or greenish; blueness of the lips or fingernail beds (cyanosis); frequent respiratory infections; lack of energy; unintended weight loss (observed in later stages of disease).
  • Subjects with COPD may also experience exacerbations, during which symptoms worsen and persist for days or longer.
  • Symptoms of pulmonary fibrosis are known in the art and include shortness of breath, particularly during exercise; dry, hacking cough; fast, shallow breathing; gradual unintended weight loss; tiredness; aching joints and muscles; and clubbing (widening and rounding of the tips of the fingers or toes).
  • Subjects suffering from COPD or pulmonary fibrosis can be identified using standard diagnostic methods routinely practiced in the art. Monitoring the effect of one or more Compounds selected from the table 1 ab and Table 1c administered to a subject who has or who is at risk of developing a pulmonary disease may be performed using the methods typically used for diagnosis.
  • pulmonary diseases or disorders that may be treated by using a compound selected from the table 1 ab and Table 1c include, for example, emphysema, asthma, bronchiectasis, and cystic fibrosis.
  • Bronchiectasis is results from damage to the airways that causes them to widen and become flabby and scarred. Bronchiectasis usually is caused by a medical condition that injures the airway walls or inhibits the airways from clearing mucus. Examples of such conditions include cystic fibrosis and primary ciliary dyskinesia (PCD).
  • PCD primary ciliary dyskinesia
  • the disorder may be caused by a blockage rather than a medical condition.
  • the methods described herein for treating or preventing (i.e., reducing the likelihood of occurrence of) a senescence associate pulmonary disease or disorder may also be used for treating a subject who is aging and has loss (or degeneration) of pulmonary function (i.e., declining or impaired pulmonary function compared with a younger subject) and/or degeneration of pulmonary tissue.
  • the respiratory system undergoes various anatomical, physiological and immunological changes with age.
  • the structural changes include chest wall and thoracic spine deformities that can impair the total respiratory system compliance resulting in increased effort to breathe.
  • the respiratory system undergoes structural, physiological, and immunological changes with age.
  • bronchoalveolar lavage An increased proportion of neutrophils and lower percentage of macrophages can be found in bronchoalveolar lavage (BAL) of older adults compared with younger adults.
  • Persistent low grade inflammation in the lower respiratory tract can cause proteolytic and oxidant-mediated injury to the lung matrix resulting in loss of alveolar unit and impaired gas exchange across the alveolar membrane seen with aging.
  • Sustained inflammation of the lower respiratory tract may predispose older adults to increased susceptibility to toxic environmental exposure and accelerated lung function decline.
  • Oxidative stress exacerbates inflammation during aging. Alterations in redox balance and increased oxidative stress during aging precipitate the expression of cytokines, chemokines, and adhesion molecules, and enzymes.
  • One or any combination of diagnostic methods including physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods described herein, may be used for monitoring the health status of the subject.
  • the effects of the treatment of a compound selected from the table 1 ab and Table 1c or pharmaceutical composition comprising the agent can be analyzed using techniques known in the art, such as comparing symptoms of patients suffering from or at risk of the pulmonary disease that have received the treatment with those of patients without such a treatment or with placebo treatment.
  • methods and techniques that evaluate mechanical functioning of the lung for example, techniques that measure lung capacitance, elastance, and airway hypersensitivity may be performed.
  • any one of numerous measurements may be obtained, expiratory reserve volume (ERV), forced vital capacity (FVC), forced expiratory volume (FEV) (e.g., FEV in one second, FEV1), FEV1/FEV ratio, forced expiratory flow 25% to 75%, and maximum voluntary ventilation (MVV), peak expiratory flow (PEF), slow vital capacity (SVC).
  • Total lung volumes include total lung capacity (TLC), vital capacity (VC), residual volume (RV), and functional residual capacity (FRC).
  • Gas exchange across alveolar capillary membrane can be measured using diffusion capacity for carbon monoxide (DLCO). Peripheral capillary oxygen saturation (SpO 2 ) can also be measured; normal oxygen levels are typically between 95% and 100%.
  • Senescence-associated diseases or disorders treatable by administering a compound selected from the table 1 ab and Table 1c described herein include neurological diseases or disorders. Such senescence-associated diseases and disorders include Parkinson's disease, Alzheimer's disease (and other dementias), motor neuron dysfunction (MND), mild cognitive impairment (MCI), Huntington's disease, and diseases and disorders of the eyes, such as age-related macular degeneration. Other diseases of the eye that are associated with increasing age are glaucoma, vision loss, presbyopia, and cataracts.
  • Parkinson's disease is the second most common neurodegenerative disease. It is a disabling condition of the brain characterized by slowness of movement (bradykinesia), shaking, stiffness, and in the later stages, loss of balance. Many of these symptoms are due to the loss of certain nerves in the brain, which results in the lack of dopamine.
  • This disease is characterized by neurodegeneration, such as the loss of about 50% to 70% of the dopaminergic neurons in the substantia nigra pars compacta, a profound loss of dopamine in the striatum, and/or the presence of intracytoplasmic inclusions (Lewy bodies), which are composed mainly of alpha-synuclein and ubiquitin.
  • Parkinson's disease also features locomotor deficits, such as tremor, rigidity, bradykinesia, and/or postural instability.
  • Subjects at risk of developing Parkinson's disease include those having a family history of Parkinson's disease and those exposed to pesticides (e.g., rotenone or paraquat), herbicides (e.g., agent orange), or heavy metals. Senescence of dopamine-producing neurons is thought to contribute to the observed cell death in PD through the production of reactive oxygen species; therefore, the methods and Compounds selected from the table 1 ab and Table 1c described herein are useful for treatment and prophylaxis of Parkinson's disease.
  • Parkinson's diseases are known in the art, such as histological studies, biochemical studies, and behavioral assessment (see, e.g., U.S. Application Publication No. 2012/0005765).
  • Symptoms of Parkinson's disease are known in the art and include, but are not limited to, difficulty starting or finishing voluntary movements, jerky, stiff movements, muscle atrophy, shaking (tremors), and changes in heart rate, but normal reflexes, bradykinesia, and postural instability.
  • people diagnosed with Parkinson's disease may have cognitive impairment, including mild cognitive impairment, in addition to their physical symptoms.
  • AD Alzheimer's disease
  • Age is the single greatest predisposing risk factor for developing AD, which is the leading cause of dementia in the elderly. Early clinical symptoms show remarkable similarity to mild cognitive impairment (see below). As the disease progresses, impaired judgment, confusion, behavioral changes, disorientation, and difficulty in walking and swallowing occur.
  • Alzheimer's disease is characterized by the presence of neurofibrillary tangles and amyloid (senile) plaques in histological specimens. The disease predominantly involves the limbic and cortical regions of the brain.
  • the argyrophilic plaques containing the amyloidogenic A ⁇ fragment of amyloid precursor protein (APP) are scattered throughout the cerebral cortex and hippocampus.
  • Neurofibrillary tangles are found in pyramidal neurons predominantly located in the neocortex, hippocampus, and nucleus basalis of Meynert.
  • Other changes such as granulovacuolar degeneration in the pyramidal cells of the hippocampus, and neuron loss and gliosis in the cortex and hippocampus, are observed.
  • Subjects at risk of developing Alzheimer's disease include those of advanced age, those with a family history of Alzheimer's disease, those with genetic risk genes (e.g., ApoE4) or deterministic gene mutations (e.g., APP, PS1, or PS2), and those with history of head trauma or heart/vascular conditions (e.g., high blood pressure, heart disease, stroke, diabetes, high cholesterol).
  • a number of behavioral and histopathological assays are known in the art for evaluating Alzheimer's disease phenotype, for characterizing therapeutic agents, and assessing treatment. Histological analyses are typically performed postmortem. Histological analysis of A ⁇ levels may be performed using Thioflavin-S.
  • Congo red or anti-A ⁇ staining (e.g., 4G8, 10D5, or 6E10 antibodies) to visualize A ⁇ deposition on sectioned brain tissues (see, e.g., Holcomb et al., 1998, Nat. Med.4:97-100; Borchelt et al., 1997, Neuron 19:939-945; Dickson et al., 1988, Am. J. Path.132:86-101).
  • In vivo methods of visualizing A ⁇ deposition in transgenic mice have been also described.
  • BSB (trans, trans)-1-bromo-2,5-bis-(3- hydroxycarbonyl-4-hydroxy)styrylbenzene) and PET tracer 11 C-labelled Pittsburgh Compound-B (PIB) bind to A ⁇ plaques (see, e.g., Skovronsky et al., 2000, Proc. Natl. Acad. Sci. USA 97:7609-7614; Klunk et al., 2004, Ann. Neurol.55:306-319).
  • F-containing amyloidophilic Congo red-type compound FSB (E,E)- 1-fluoro-2,5-bis-(3-hydroxycarbonyl-4-hydroxy)styrylbenzene) allows visualization of A ⁇ plaques by MRI (see, e.g., Higuchi et al., 2005, Nature Neurosci.8:527-533). Radiolabeled, putrescine-modified amyloid- beta peptide labels amyloid deposits in vivo in a mouse model of Alzheimer's disease (see, e.g., Wengenack et al., 2000, Nat. Biotechnol.18:868-872).
  • GFAP glial fibrillary acidic protein
  • Neurofibrillary tangles may be identified by immunohistochemistry using thioflavin-S fluorescent microscopy and Gallyas silver stains (see, e.g., Gotz et al., 2001, J. Biol. Chem.276:529-534; U.S. Pat. No. 6,664,443). Axon staining with electron microscopy and axonal transport studies may be used to neuronal degeneration (see, e.g., Ishihara et al., 1999, Neuron 24:751-762). [00112] Subjects suffering from Alzheimer's disease can be identified using standard diagnostic methods known in the art for Alzheimer's disease.
  • Alzheimer's disease is based on symptoms (e.g., progressive decline in memory function, gradual retreat from and frustration with normal activities, apathy, agitation or irritability, aggression, anxiety, sleep disturbance, dysphoria, aberrant motor behavior, disinhibition, social withdrawal, decreased appetite, hallucinations, dementia), medical history, neuropsychological tests, neurological and/or physical examination of a patient. Cerebrospinal fluid may also be for tested for various proteins that have been associated with Alzheimer pathology, including tau, amyloid beta peptide, and AD7C-NTP. Genetic testing is also available for early-onset familial Alzheimer disease (eFAD), an autosomal-dominant genetic disease.
  • eFAD early-onset familial Alzheimer disease
  • MCI Mild Cognitive Impairment
  • MCI that primarily affects memory is known as “amnestic MCI.”
  • Amnestic MCI A person with amnestic MCI may start to forget important information that he or she would previously have recalled easily, such as recent events. Amnestic MCI is frequently seen as prodromal stage of Alzheimer's disease. MCI that affects thinking skills other than memory is known as “non-amnestic MCI.” This type of MCI affect thinking skills such as the ability to make sound decisions, judge the time or sequence of steps needed to complete a complex task, or visual perception. Individuals with non-amnestic MCI are believed to be more likely to convert to other types of dementias (e.g., dementia with Lewy bodies).
  • MND Motor Neuron Dysfunction
  • MNDs include, but are not limited to Amyotrophic Lateral Sclerosis (ALS), also known as Lou Gehrig's Disease, progressive bulbar palsy, pseudobulbar palsy, primary lateral sclerosis, progressive muscular atrophy, lower motor neuron disease, and spinal muscular atrophy (SMA) (e.g., SMA1 also called Werdnig-Hoffmann Disease, SMA2, SMA3 also called Kugelberg-Welander Disease, and Kennedy's disease), post-polio syndrome, and hereditary spastic paraplegia.
  • ALS Amyotrophic Lateral Sclerosis
  • SMA spinal muscular atrophy
  • SMA1 also called Werdnig-Hoffmann Disease
  • SMA2 SMA3 also called Kugelberg-Welander Disease
  • Kennedy's disease post-polio syndrome
  • hereditary spastic paraplegia hereditary spastic paraplegia.
  • ALS amyotrophic lateral sclerosis
  • ALS amyotrophic lateral sclerosis
  • Primary lateral sclerosis is a disease of the upper motor neurons, while progressive muscular atrophy affects only lower motor neurons in the spinal cord. In progressive bulbar palsy, the lowest motor neurons of the brain stem are most affected, causing slurred speech and difficulty chewing and swallowing. There are almost always mildly abnormal signs in the arms and legs.
  • Patients with MND exhibit a phenotype of Parkinson's disease (e.g., having tremor, rigidity, bradykinesia, and/or postural instability).
  • Methods for detecting, monitoring or quantifying locomotor and/or other deficits associated with Parkinson's diseases, such as MND are known in the art (see, e.g., U.S. Application Publication No.20120005765).
  • MNDs are characterized by death of motor neurons, progressive accumulation of detergent-resistant aggregates containing SOD1 and ubiquitin and aberrant neurofilament accumulations in degenerating motor neurons.
  • reactive astroglia and microglia are often detected in diseased tissue.
  • a senescence-associated disease or disorder is an ocular disease, disorder, or condition, for example, presbyopia, macular degeneration, or cataracts.
  • the senescence-associated disease or disorder is glaucoma.
  • Macular degeneration is a neurodegenerative disease that causes the loss of photoreceptor cells in the central part of retina, called the macula. Macular degeneration generally is classified into two types: dry type and wet type.
  • the dry form is more common than the wet, with about 90% of age-related macular degeneration (ARMD or AMD) patients diagnosed with the dry form.
  • ARMD age-related macular degeneration
  • AMD age-related macular degeneration
  • the wet form of the disease usually leads to more serious vision loss. While the exact causes of age-related macular degeneration are still unknown, the number of senescent retinal pigmented epithelial (RPE) cells increases with age.
  • Age and certain genetic factors and environmental factors are risk factors for developing ARMD. Environment predisposing factors include omega-3 fatty acids intake; estrogen exposure; and increased serum levels of vitamin D.
  • Dicer1 enzyme involved in maturation of micro RNA
  • DICER1 ablation induces premature senescence.
  • Dry ARMD is associated with atrophy of RPE layer, which causes loss of photoreceptor cells. The dry form of ARMD may result from aging and thinning of macular tissues and from deposition of pigment in the macula. Senescence appears to inhibit both replication and migration of RPE, resulting in permanent RPE depletion in the macula of dry AMD patients. With wet ARMD, new blood vessels grow beneath the retina and leak blood and fluid.
  • This abnormal leaky choroidal neovascularization causes the retinal cells to die, creating blind spots in central vision.
  • Different forms of macular degeneration may also occur in younger patients. Non-age related etiology may be linked to heredity, diabetes, nutritional deficits, head injury, infection, or other factors.
  • Declining vision noticed by the patient or by an ophthalmologist during a routine eye exam may be the first indicator of macular degeneration.
  • the formation of exudates, or “drusen,” underneath the Bruch's membrane of the macula is often the first physical sign that macular degeneration may develop.
  • Symptoms include perceived distortion of straight lines and, in some cases, the center of vision appears more distorted than the rest of a scene; a dark, blurry area or “white-out” appears in the center of vision; and/or color perception changes or diminishes. Diagnosing and monitoring of a subject with macular degeneration may be accomplished by a person skilled in the ophthalmic art according to art-accepted periodic eye examination procedures and report of symptoms by the subject. [00125] Presbyopia is an age-related condition where the eye exhibits a progressively diminished ability to focus on near objects as the speed and amplitude of accommodation of a normal eye decreases with advancing age. Loss of elasticity of the crystalline lens and loss of contractility of the ciliary muscles have been postulated as its cause.
  • the laminated structure of the capsule also changes and may result, at least in part, from a change in the composition of the tissue.
  • the major structural component of the lens capsule is basement membrane type IV collagen that is organized into a three-dimensional molecular network.
  • Type IV collagen is composed of six homologous ⁇ chains ( ⁇ 1-6) that associate into heterotrimeric collagen IV protomers with each comprising a specific chain combination of ⁇ 112, ⁇ 345, or ⁇ 556.
  • Protomers share structural similarities of a triple-helical collagenous domain with the triplet peptide sequence of Gly-X-Y, ending in a globular C-terminal region termed the non-collagenous 1 (NC1) domain.
  • the N-termini are composed of a helical domain termed the 7S domain, which is also involved in protomer-protomer interactions.
  • PCO Posterior capsule opacification
  • PCO results from proliferation and activity of residual lens epithelial cells along the posterior capsule in a response akin to wound healing.
  • Growth factors such as fibroblast growth factor, transforming growth factor ⁇ , epidermal growth factor, hepatocyte growth factor, insulin-like growth factor, and interleukins IL-1 and IL-6 may also promote epithelial cell migration.
  • production of these factors and cytokines by senescent cells contribute to the SASP.
  • collagen IV promotes adherence of lens epithelial cells. Adhesion of the collagen IV, fibronectin, and laminin to the intraocular lens inhibits cell migration and may reduce the risk of PCO.
  • selective killing of senescent cells by the and/or other properties of Compounds selected from the table 1 ab and Table 1c may slow or impede (delay, inhibit, retard) the disorganization of the type IV collagen network. Removal of senescence cells and thereby removing the inflammatory effects of SASP may decrease or inhibit epithelial cell migration and may also delay (suppress) the onset of presbyopia or decrease or slow the progressive severity of the condition (such as slow the advancement from mild to moderate or moderate to severe).
  • the Compounds selected from the table 1 ab and Table 1c described herein may also be useful for post-cataract surgery to reduce the likelihood of occurrence of PCO.
  • At least one compound selected from the table 1 ab and Table 1c that selectively kills senescent cells may be administered to a subject who is at risk of developing presbyopia, cataracts, or macular degeneration.
  • Treatment with a compound selected from the table 1 ab and Table 1c may be initiated when a human subject is at least 40 years of age to delay or inhibit onset or development of cataracts, presbyopia, and macular degeneration. Because almost all humans develop presbyopia, in certain embodiments, the compound selected from the table 1 ab and Table 1c may be administered in a manner as described herein to a human subject after the subject reaches the age of 40 to delay or inhibit onset or development of presbyopia.
  • the senescence associated disease or disorder is glaucoma.
  • Glaucoma is a broad term used to describe a group of diseases that causes visual field loss, often without any other prevailing symptoms. The lack of symptoms often leads to a delayed diagnosis of glaucoma until the terminal stages of the disease. Even if subjects afflicted with glaucoma do not become blind, their vision is often severely impaired. Normally, clear fluid flows into and out of the front part of the eye, known as the anterior chamber. In individuals who have open/wide-angle glaucoma, this fluid drains too slowly, leading to increased pressure within the eye. If left untreated, this high pressure subsequently damages the optic nerve and can lead to complete blindness.
  • ganglion cells are a specific type of projection neuron that connects the eye to the brain.
  • SA- ⁇ -Gal staining When the cellular network required for the outflow of fluid was subjected to SA- ⁇ -Gal staining, a fourfold increase in senescence has been observed in glaucoma patients.
  • standard automated perimetry visual field test
  • several algorithms for progression detection have been developed (see, e.g., Wesselink et al., Arch Ophthalmol.127(3):270- 274 (2009), and references therein).
  • Additional methods include gonioscopy (examines the trabecular meshwork and the angle where fluid drains out of the eye); imaging technology, for example scanning laser tomography (e.g., HRT3), laser polarimetry (e.g., GDX), and ocular coherence tomography); ophthalmoscopy; and pachymeter measurements that determine central corneal thickness.
  • gonioscopy examines the trabecular meshwork and the angle where fluid drains out of the eye
  • imaging technology for example scanning laser tomography (e.g., HRT3), laser polarimetry (e.g., GDX), and ocular coherence tomography); ophthalmoscopy; and pachymeter measurements that determine central corneal thickness.
  • gonioscopy examines the trabecular meshwork and the angle where fluid drains out of the eye
  • imaging technology for example scanning laser tomography (e.g., HRT3), laser polarimetry (e.g., GDX
  • Diabetes is characterized by high levels of blood glucose caused by defects in insulin production, insulin action, or both. The great majority (90 to 95%) of all diagnosed cases of diabetes in adults are type 2 diabetes, characterized by the gradual loss of insulin production by the pancreas. Diabetes is the leading cause of kidney failure, nontraumatic lower-limb amputations, and new cases of blindness among adults in the U.S. Diabetes is a major cause of heart disease and stroke and is the seventh leading cause of death in the U.S. (see, e.g., Centers for Disease Control and Prevention, National diabetes fact sheet: national estimates and general information on diabetes and pre-diabetes in the United States, 2011 (“Diabetes fact sheet”)).
  • Compounds selected from the table 1 ab and Table 1c described herein may be used for treating type 2 diabetes, particularly age-, diet- and obesity-associated type 2 diabetes.
  • Involvement of senescent cells in metabolic disease, such as obesity and type 2 diabetes has been suggested as a response to injury or metabolic dysfunction.
  • Fat tissue from obese mice showed induction of the senescence markers SA- ⁇ -Gal, p53, and p21.
  • a concomitant up-regulation of pro- inflammatory cytokines, such as tumor necrosis factor- ⁇ and Ccl2/MCP1 was observed in the same fat tissue.
  • Induction of senescent cells in obesity potentially has clinical implications because pro- inflammatory SASP components are also suggested to contribute to type 2 diabetes.
  • a similar pattern of up-regulation of senescence markers and SASP components are associated with diabetes, both in mice and in humans. Accordingly, the methods described herein that comprise administering a compound selected from the table 1 ab and Table 1c may be useful for treatment or prophylaxis of type 2 diabetes, as well as obesity and metabolic syndrome. Without wishing to be bound by theory, contact of senescent pre- adipocytes with a compound selected from the table 1 ab and Table 1c thereby killing the senescent pre- adipocytes may provide clinical and health benefit to a person who has any one of diabetes, obesity, or metabolic syndrome. [00137] Subjects suffering from type 2 diabetes can be identified using standard diagnostic methods known in the art for type 2 diabetes.
  • diagnosis of type 2 diabetes is based on symptoms (e.g., increased thirst and frequent urination, increased hunger, weight loss, fatigue, blurred vision, slow-healing sores or frequent infections, and/or areas of darkened skin), medical history, and/or physical examination of a patient.
  • Subjects at risk of developing type 2 diabetes include those who have a family history of type 2 diabetes and those who have other risk factors such as excess weight, fat distribution, inactivity, race, age, prediabetes, and/or gestational diabetes.
  • the effectiveness of a compound selected from the table 1 ab and Table 1c can readily be determined by a person skilled in the medical and clinical arts.
  • One or any combination of diagnostic methods including physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods, such as those described herein, may be used for monitoring the health status of the subject.
  • a subject who is receiving one or more Compounds selected from the table 1 ab and Table 1c described herein for treatment or prophylaxis of diabetes can be monitored, for example, by assaying glucose and insulin tolerance, energy expenditure, body composition, fat tissue, skeletal muscle, and liver inflammation, and/or lipotoxicity (muscle and liver lipid by imaging in vivo and muscle, liver, bone marrow, and pancreatic ⁇ -cell lipid accumulation and inflammation by histology).
  • BMI Body Mass index
  • obesity and obesity-related refer to human subjects with body mass index values of greater than 30, greater than 35, or greater than 40.
  • a category of obesity not captured by BMI is called “abdominal obesity” in the art, which relates to the extra fat found around a subject's middle, which is an important factor in health, even independent of BMI.
  • the simplest and most often used measure of abdominal obesity is waist size.
  • abdominal obesity in women is defined as a waist size 35 inches or higher, and in men as a waist size of 40 inches or higher.
  • More complex methods for determining obesity require specialized equipment, such as magnetic resonance imaging or dual energy X-ray absorptioimetry machines.
  • a condition or disorder associated with diabetes and senescence is a diabetic ulcer (i.e., diabetic wound).
  • An ulcer is a breakdown in the skin, which may extend to involve the subcutaneous tissue or even muscle or bone. These lesions occur, particularly, on the lower extremities.
  • Patients with diabetic venous ulcer exhibit elevated presence of cellular senescence at sites of chronic wounds. Chronic inflammation is also observed at sites of chronic wounds, such as diabetic ulcers, suggesting that the proinflammatory cytokine phenotype of senescent cells has a role in the pathology.
  • Subjects who have type 2 diabetes or who are at risk of developing type 2 diabetes may have metabolic syndrome.
  • Metabolic syndrome in humans is typically associated with obesity and characterized by one or more of cardiovascular disease, liver steatosis, hyperlipidemia, diabetes, and insulin resistance.
  • a subject with metabolic syndrome may present with a cluster of metabolic disorders or abnormalities which may include, for example, one or more of hypertension, type-2 diabetes, hyperlipidemia, dyslipidemia (e.g., hypertriglyceridemia, hypercholesterolemia), insulin resistance, liver steatosis (steatohepatitis), hypertension, atherosclerosis, and other metabolic disorders.
  • Renal Dysfunction [00143] Nephrological pathologies, such as glomerular disease, arise in the elderly.
  • Glomerulonephritis is characterized by inflammation of the kidney and by the expression of two proteins, IL1 ⁇ and IL1 ⁇ . Glomerular disease is associated with elevated presence of senescent cells, especially in fibrotic kidneys.
  • Dermatological Disease or Disorder include dermatological diseases or disorders. Such senescent cell associated diseases and disorders include psoriasis and eczema, which are also inflammatory diseases and are discussed in greater detail above.
  • Other dermatological diseases and disorders that are associated with senescence include rhytides (wrinkles due to aging); pruritis (linked to diabetes and aging); dysesthesia (chemotherapy side effect that is linked to diabetes and multiple sclerosis); psoriasis (as noted) and other papulosquamous disorders, for example, erythroderma, lichen planus, and lichenoid dermatosis; atopic dermatitis (a form of eczema and associated with inflammation); eczematous eruptions (often observed in aging patients and linked to side effects of certain drugs).
  • Other dermatological diseases and disorders associated with senescence include eosinophilic dermatosis (linked to certain kinds of hemotologic cancers); reactive neutrophilic dermatosis (associated with underlying diseases such as inflammatory bowel syndrome); pemphigus (an autoimmune disease in which autoantibodies form against desmoglein); pemphigoid and other immunobullous dermatosis (autoimmune blistering of skin); fibrohistocytic proliferations of skin, which is linked to aging; and cutaneous lymphomas that are more common in older populations.
  • Another dermatological disease that may be treatable according to the methods described herein includes cutaneous lupus, which is a symptom of lupus erythematosus.
  • Late onset lupus may be linked to decreased (i.e., reduced) function of T-cell and B-cells and cytokines (immunosenescence) associated with aging.
  • cytokines immunosenescence
  • Metastasis In a particular embodiment, methods are provided for treating or preventing (i.e., reducing the likelihood of occurrence or development of) a senescence cell associated disease (or disorder or condition), which is metastasis.
  • the Compounds selected from the table 1 ab and Table 1c described herein may also be used according to the methods described herein for treating or preventing (i.e., reducing the likelihood of occurrence of) metastasis (i.e., the spreading and dissemination of cancer or tumor cells) from one organ or tissue to another organ or tissue in the body.
  • a senescent cell-associated disease or disorder includes metastasis, and a subject who has a cancer may benefit from administration of a compound selected from the table 1 ab and Table 1c as described herein for inhibiting metastasis.
  • Such a compound selected from the table 1 ab and Table 1c when administered to a subject who has a cancer according to the methods described herein may inhibit tumor proliferation.
  • Metastasis of a cancer occurs when the cancer cells (i.e., tumor cells) spread beyond the anatomical site of origin and initial colonization to other areas throughout the body of the subject.
  • Tumor proliferation may be determined by tumor size, which can be measured in various ways familiar to a person skilled in the art, such as by PET scanning, MRI, CAT scan, biopsy, for example.
  • the effect of the therapeutic agent on tumor proliferation may also be evaluated by examining differentiation of the tumor cells.
  • cancer or tumor are clinically descriptive terms that encompass diseases typically characterized by cells exhibiting abnormal cellular proliferation.
  • the term cancer is generally used to describe a malignant tumor or the disease state arising from the tumor.
  • an abnormal growth may be referred to in the art as a neoplasm.
  • the term tumor such as in reference to a tissue, generally refers to any abnormal tissue growth that is characterized, at least in part, by excessive and abnormal cellular proliferation.
  • a tumor may be metastatic and capable of spreading beyond its anatomical site of origin and initial colonization to other areas throughout the body of the subject.
  • a cancer may comprise a solid tumor or may comprise a “liquid” tumor (e.g., leukemia and other blood cancers).
  • Cells are induced to senesce by cancer therapies, such as radiation and certain chemotherapy drugs.
  • senescent cells increases secretion of inflammatory molecules (see description herein of senescent cells), promotes tumor progression, which may include promoting tumor growth and increasing tumor size, promoting metastasis, and altering differentiation.
  • tumor progression is significantly inhibited, resulting in tumors of small size and with little or no observed metastatic growth (see, e.g., Int'l Appl. Publication No. WO 2013/090645).
  • methods are provided for preventing (i.e., reducing the likelihood of occurrence of), inhibiting, or retarding metastasis in a subject who has a cancer by administering a compound selected from the table 1 ab and Table 1c as described herein.
  • the compound selected from the table 1 ab and Table 1c is administered on one or more days within a treatment window (i.e., treatment course) of no longer than 7 days or 14 days.
  • a treatment window i.e., treatment course
  • the treatment course is no longer than 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or no longer than 21 days.
  • the treatment course is a single day.
  • the compound selected from the table 1 ab and Table 1c is administered on two or more days within a treatment window of no longer than 7 days or 14 days, on 3 or more days within a treatment window of no longer than 7 days or 14 days; on 4 or more days within a treatment window of no longer than 7 days or 14 days; on 5 or more days within a treatment window of no longer than 7 days or 14 days; on 6, 7, 8, 9, 10, 11, 12, 13, or 14 days within treatment window of no longer than 7 days or 14 days.
  • the agent may be administered every 2 nd day (i.e., every other day).
  • the agent when the at least one compound selected from the table 1 ab and Table 1c is administered to a subject for a treatment window of 4 days or more, the agent may be administered every 3 rd day (i.e., every other third day).
  • cancer therapies such as radiation and certain chemotherapy drugs (e.g., doxorubicin; paclitaxel; gemcitabine; pomalidomide; lenalidomide)
  • a compound selected from the table 1 ab and Table 1c described herein may be administered after the chemotherapy or radiotherapy to kill (or facilitate killing) of these senescent cells.
  • senescence-associated secretory phenotype SASP
  • the following treatment courses for administration of the compound selected from the table 1 ab and Table 1c may be used in methods described herein for treating or preventing (i.e., reducing the likelihood of occurrence, or reducing the severity) a chemotherapy or radiotherapy side effect.
  • the compound selected from the table 1 ab and Table 1c is administered on one or more days during the off-therapy time interval (time period) beginning on or after the second day of the off-therapy time interval and ending on or before the last day of the off-therapy time interval.
  • n is the number of days off- therapy
  • the compound selected from the table 1 ab and Table 1c is administered on at least one day and no more than n ⁇ 1 days of the off-therapy time interval.
  • chemotherapy or radiotherapy is administered in a treatment cycle of at least one day on-therapy (i.e., chemotherapy or radiotherapy)) followed by at least one week off-therapy
  • the compound selected from the table 1 ab and Table 1c is administered on one or more days during the off-therapy time interval beginning on or after the second day of the off-therapy time interval and ending on or before the last day of the off- therapy time interval.
  • the compound selected from the table 1 ab and Table 1c is administered on one day that is the sixth day of the off-therapy time interval.
  • chemotherapy or radiotherapy is administered in a treatment cycle of at least one day on-therapy (i.e., chemotherapy or radiotherapy)) followed by at least two weeks off-therapy
  • the compound selected from the table 1 ab and Table 1c is administered beginning on the sixth day of the off-chemo- or radio-therapy time interval and ending at least one day or at least two days prior to the first day of a subsequent chemotherapy or radiation therapy treatment course.
  • a compound selected from the table 1 ab and Table 1c may be administered on at least one and on no more than 7 days (i.e., 1, 2, 3, 4, 5, 6, or 7 days) of the off-therapy time interval beginning on the sixth day after the chemotherapy or radiotherapy course ends (i.e., the sixth day of the off chemo-radio-therapy interval).
  • a compound selected from the table 1 ab and Table 1c may be administered on at least one day and on no more than 14 days (i.e., 1-14 days: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days) of the off-therapy time interval beginning on the sixth day after the chemotherapy or radiotherapy course ends.
  • the compound selected from the table 1 ab and Table 1c treatment course is at least one day and no longer than 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or no more than 21 days (i.e., 1-21 days), provided that administration of the compound selected from the table 1 ab and Table 1c is not concurrent with the chemotherapy or radiotherapy.
  • the compound selected from the table 1 ab and Table 1c treatment course is a single day.
  • the compound selected from the table 1 ab and Table 1c is administered on two or more days within a treatment window of no longer than 14 days, on 3 or more days within a treatment window of no longer than 14 days; on 4 or more days within a treatment window of no longer than 14 days; on 5 or more days within a treatment window of no longer than 14 days; on 6, 7, 8, 9, 10, 11, 12, 13, or 14 days within treatment window of no longer than 14 days.
  • the agent may be administered every 2 nd day (i.e., every other day).
  • the agent when the at least one compound selected from the table 1 ab and Table 1c is administered to a subject during a treatment course of 4 days or more, the agent may be administered every 3 rd day (i.e., every other third day).
  • Many chemotherapy and radiotherapy treatment regimens comprise a finite number of cycles of on-drug therapy followed by off-drug therapy or comprise a finite timeframe in which the chemotherapy or radiotherapy is administered.
  • Such cancer treatment regimens may also be called treatment protocols. The protocols are determined by clinical trials, drug labels, and clinical staff in conjunction with the subject to be treated.
  • the number of cycles of a chemotherapy or radiotherapy or the total length of time of a chemotherapy or radiotherapy regimen can vary depending on the patient's response to the cancer therapy.
  • a compound selected from the table 1 ab and Table 1c may be administered after the treatment regimen of chemotherapy or radiotherapy has been completed.
  • the compound selected from the table 1 ab and Table 1c is administered after the chemotherapy or radiotherapy has been completed on one or more days within treatment window (i.e., compound selected from the table 1 ab and Table 1c treatment course) of no longer than 14 days.
  • the compound selected from the table 1 ab and Table 1c treatment course is no longer than 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or no more than 21 days.
  • the treatment course is a single day.
  • the compound selected from the table 1 ab and Table 1c is administered on two or more days within a treatment window of no longer than 14 days, on 3 or more days within a treatment window of no longer than 14 days; on 4 or more days within a treatment window of no longer than 14 days; on 5 or more days within a treatment window of no longer than 14 days; on 6, 7, 8, 9, 10, 11, 12, 13, or 14 days within treatment window of no longer than 14 days.
  • the agent when the at least one compound selected from the table 1 ab and Table 1c is administered to a subject after chemotherapy or radiotherapy for a treatment window of 3 days or more, the agent may be administered every 2 nd day (i.e., every other day).
  • the agent when the at least one compound selected from the table 1 ab and Table 1c is administered to a subject for a treatment window of 4 days or more, the agent may be administered every 3 rd d day (i.e., every other third day).
  • the treatment with the compound selected from the table 1 ab and Table 1c may be initiated at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days or later after the cancer treatment regimen has been completed.
  • the treatment with the compound selected from the table 1 ab and Table 1c may be initiated at least 6, 7, 8, 9, 10, 11, 12, 13, or 14 days or later after the cancer treatment regimen has been completed.
  • a chemotherapy may be referred to as a chemotherapy, chemotherapeutic, or chemotherapeutic drug.
  • Many chemotherapeutics are compounds referred to as small organic molecules.
  • Chemotherapy is a term that is also used to describe a combination chemotherapeutic drugs that are administered to treat a particular cancer.
  • a chemotherapy may also refer to a combination of two or more chemotherapeutic molecules that are administered coordinately and which may be referred to as combination chemotherapy.
  • a cancer that may metastasize may be a solid tumor or may be a liquid tumor (e.g., a blood cancer, for example, a leukemia).
  • a liquid tumor e.g., a blood cancer, for example, a leukemia.
  • Cancers that are liquid tumors are classified in the art as those that occur in blood, bone marrow, and lymph nodes and include generally, leukemias (myeloid and lymphocytic), lymphomas (e.g., Hodgkin lymphoma), and melanoma (including multiple myeloma).
  • Leukemias include for example, acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), and hairy cell leukemia.
  • ALL acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • hairy cell leukemia hairy cell leukemia.
  • Cancers that are solid tumors and occur in greater frequency in humans include, for example, prostate cancer, testicular cancer, breast cancer, brain cancer, pancreatic cancer, colon cancer, thyroid cancer, stomach cancer, lung cancer, ovarian cancer, Kaposi's sarcoma, skin cancer (including squamous cell skin cancer), renal cancer, head and neck cancers, throat cancer, squamous carcinomas that form on the moist mucosal linings of the nose, mouth, throat, etc.), bladder cancer, osteosarcoma (bone cancer), cervical cancer, endometrial cancer, esophageal cancer, liver cancer, and kidney cancer.
  • prostate cancer testicular cancer
  • breast cancer brain cancer
  • pancreatic cancer colon cancer
  • thyroid cancer stomach cancer
  • lung cancer ovarian cancer
  • Kaposi's sarcoma skin cancer (including squamous cell skin cancer), renal cancer, head and neck cancers, throat cancer, squamous carcinomas that form on the moist mucosal linings of the nose, mouth
  • the senescent cell- associated disease or disorder treated or prevented (i.e., likelihood of occurrence or development is reduced) by the methods described herein is metastasis of melanoma cells, prostate cancer cells, testicular cancer cells, breast cancer cells, brain cancer cells, pancreatic cancer cells, colon cancer cells, thyroid cancer cells, stomach cancer cells, lung cancer cells, ovarian cancer cells, Kaposi's sarcoma cells, skin cancer cells, renal cancer cells, head or neck cancer cells, throat cancer cells, squamous carcinoma cells, bladder cancer cells, osteosarcoma cells, cervical cancer cells, endometrial cancer cells, esophageal cancer cells, liver cancer cells, or kidney cancer cells.
  • Types of cancers include the following: adrenocortical carcinoma, childhood adrenocortical carcinoma, aids-related cancers, anal cancer, appendix cancer, basal cell carcinoma, childhood basal cell carcinoma, bladder cancer, childhood bladder cancer, bone cancer, brain tumor, childhood astrocytomas, childhood brain stem glioma, childhood central nervous system atypical teratoid/rhabdoid tumor, childhood central nervous system embryonal tumors, childhood central nervous system germ cell tumors, childhood craniopharyngioma brain tumor, childhood ependymoma brain tumor, breast cancer, childhood bronchial tumors, carcinoid tumor, childhood carcinoid tumor, gastrointestinal carcinoid tumor, carcinoma of unknown primary, childhood carcinoma of unknown primary, childhood cardiac (heart) tumors, cervical cancer, childhood cervical cancer
  • the senescence cell associated disorder or condition is a chemotherapeutic side effect or a radiotherapy side effect.
  • chemotherapeutic agents that include non-cancer cells to senesce include anthracyclines (such as doxorubicin, daunorubicin); taxols (e.g., paclitaxel); gemcitabine; pomalidomide; and lenalidomide.
  • anthracyclines such as doxorubicin, daunorubicin
  • taxols e.g., paclitaxel
  • gemcitabine pomalidomide
  • lenalidomide pomalidomide
  • lenalidomide lenalidomide
  • One or more of the Compounds selected from the table 1 ab and Table 1c administered as described herein may be used for treating and/or preventing (i.e., reducing the likelihood of occurrence of) a chemotherapeutic side effect or a radiotherapy side effect.
  • Acute toxic side effects include but are not limited to gastrointestinal toxicity (e.g., nausea, vomiting, constipation, anorexia, diarrhea), peripheral neuropathy, fatigue, malaise, low physical activity, hematological toxicity (e.g., anemia), hepatotoxicity, alopecia (hair loss), pain, infection, mucositis, fluid retention, dermatological toxicity (e.g., rashes, dermatitis, hyperpigmentation, urticaria, photosensitivity, nail changes), mouth (e.g., oral mucositis), gum or throat problems, or any toxic side effect caused by a chemotherapy or radiotherapy.
  • gastrointestinal toxicity e.g., nausea, vomiting, constipation, anorexia, diarrhea
  • peripheral neuropathy e.g., fatigue, malaise, low physical activity
  • hematological toxicity e.g., anemia
  • hepatotoxicity e.g., hepatotoxicity
  • alopecia hair loss
  • pain infection
  • toxic side effects caused by radiotherapy or chemotherapy may be ameliorated by the methods described herein.
  • methods are provided herein for ameliorating (reducing, inhibiting, or preventing occurrence (i.e., reducing the likelihood of occurrence)) acute toxicity or reducing severity of a toxic side effect (i.e., deleterious side effect) of a chemotherapy or radiotherapy or both in a subject who receives the therapy, wherein the method comprises administering to the subject an agent that selectively kills, removes, or destroys or facilitates selective destruction of senescent cells.
  • Administration of a compound selected from the table 1 ab and Table 1c for treating or reducing the likelihood of occurrence, or reducing the severity of a chemotherapy or radiotherapy side effect may be accomplished by the same treatment courses described above for treatment/prevention of metastasis.
  • the compound selected from the table 1 ab and Table 1c is administered during the off- chemotherapy or off-radiotherapy time interval or after the chemotherapy or radiotherapy treatment regimen has been completed.
  • the acute toxicity is an acute toxicity comprising energy imbalance and may comprise one or more of weight loss, endocrine change(s) (e.g., hormone imbalance, change in hormone signaling), and change(s) in body composition.
  • an acute toxicity comprising energy imbalance relates to decreased or reduced ability of the subject to be physically active, as indicated by decreased or diminished expenditure of energy than would be observed in a subject who did not receive the medical therapy.
  • such an acute toxic effect that comprises energy imbalance includes low physical activity.
  • energy imbalance comprises fatigue or malaise.
  • a chemotherapy side effect to be treated or prevented (i.e., likelihood of occurrence is reduced) by a compound selected from the table 1 ab and Table 1c is cardiotoxicity.
  • a subject who has a cancer that is being treated with an anthracycline such as doxorubicin, daunorubicin
  • anthracycline such as doxorubicin, daunorubicin
  • the maximum lifetime dose that a subject can receive is limited even if the cancer is responsive to the drug.
  • Administration of one or more of the Compounds selected from the table 1 ab and Table 1c may reduce the cardiotoxicity such that additional amounts of the anthracycline can be administered to the subject, resulting in an improved prognosis related to cancer disease.
  • the cardiotoxicity results from administration of an anthracyline, such as doxorubicin.
  • Doxorubicin is an anthracycline topoisomerase that is approved for treating patients who have ovarian cancer after failure of a platinum based therapy; Kaposi's sarcoma after failure of primary systemic chemotherapy or intolerance to the therapy; or multiple myeloma in combination with bortezomib in patients who have not previously received bortezomib or who have received at least one prior therapy.
  • Doxorubicin may cause myocardial damage that could lead to congestive heart failure if the total lifetime dose to a patient exceeds 550 mg/m 2 . Cardiotoxicity may occur at even lower doses if the patient also receives mediastinal irradiation or another cardiotoxic drug.
  • a compound selected from the table 1 ab and Table 1c may be used in the methods as provided herein for ameliorating chronic or long term side effects.
  • Chronic toxic side effects typically result from multiple exposures to or administrations of a chemotherapy or radiotherapy over a longer period of time.
  • Certain toxic effects appear long after treatment also called late toxic effects
  • Organ dysfunction e.g., neurological, pulmonary, cardiovascular, and endocrine dysfunction
  • Chronic and/or late toxic side effects that occur in subjects who received chemotherapy or radiation therapy include by way of non-limiting example, cardiomyopathy, congestive heart disease, inflammation, early menopause, osteoporosis, infertility, impaired cognitive function, peripheral neuropathy, secondary cancers, cataracts and other vision problems, hearing loss, chronic fatigue, reduced lung capacity, and lung disease.
  • the sensitivity to the chemotherapy or the radiotherapy may be enhanced in a clinically or statistically significant manner than if the compound selected from the table 1 ab and Table 1c was not administered.
  • development of chemotherapy or radiotherapy resistance may be inhibited when a compound selected from the table 1 ab and Table 1c is administered to a subject treated with the respective chemotherapy or radiotherapy.
  • a compound selected from the table 1 ab and Table 1c may also be useful for treating or preventing (i.e., reducing the likelihood of occurrence) of an age-related disease or disorder that occurs as part of the natural aging process or that occurs when the subject is exposed to a senescence inducing agent or factor (e.g., irradiation, chemotherapy, smoking tobacco, high-fat/high sugar diet, other environmental factors).
  • a senescence inducing agent or factor e.g., irradiation, chemotherapy, smoking tobacco, high-fat/high sugar diet, other environmental factors.
  • An age-related disorder or disease or an age-sensitive trait may be associated with a senescence- inducing stimulus.
  • the efficacy of a method of treatment described herein may be manifested by reducing the number of symptoms of an age-related disorder or age-sensitive trait associated with a senescence- inducing stimulus, decreasing the severity of one or more symptoms, or delaying the progression of an age- related disorder or age-sensitive trait associated with a senescence-inducing stimulus.
  • preventing an age-related disorder or age-sensitive trait associated with a senescence- inducing stimulus refers to preventing (i.e., reducing the likelihood of occurrence) or delaying onset of an age-related disorder or age-sensitive trait associated with a senescence-inducing stimulus, or reoccurrence of one or more age-related disorder or age-sensitive trait associated with a senescence-inducing stimulus.
  • Age related diseases or conditions include, for example, renal dysfunction, kyphosis, herniated intervertebral disc, frailty, hair loss, hearing loss, vision loss (blindness or impaired vision), muscle fatigue, skin conditions, skin nevi, diabetes, metabolic syndrome, and sarcopenia.
  • Vision loss refers to the absence of vision when a subject previously had vision.
  • Various scales have been developed to describe the extent of vision and vision loss based on visual acuity.
  • Age-related diseases and conditions also include dermatological conditions, for example without limitation, treating one or more of the following conditions: wrinkles, including superficial fine wrinkles; hyperpigmentation; scars; keloid; dermatitis; psoriasis; eczema (including seborrheic eczema); rosacea; vitiligo; ichthyosis vulgaris; dermatomyositis; and actinic keratosis.
  • frailty is defined as a clinically recognizable state of increased vulnerability resulting from aging-associated decline in reserve and function across multiple physiologic systems that compromise a subject's ability to cope with every day or acute stressors. Frailty has been may be characterized by compromised energetics characteristics such as low grip strength, low energy, slowed waking speed, low physical activity, and/or unintentional weight loss. Studies have suggested that a patient may be diagnosed with frailty when three of five of the foregoing characteristics are observed (see, e.g., Fried et al., J. Gerontol. A Biol. Sci. Med, Sci.2001; 56(3):M146-M156; Xue, Clin. Geriatr.
  • aging and diseases and disorders related to aging may be treated or prevented (i.e., the likelihood of occurrence of is reduced) by administering a Compound selected from the table 1 ab and Table 1c .
  • the compound selected from the table 1 ab and Table 1c may inhibit senescence of adult stem cells or inhibit accumulation, kill, or facilitate removal of adult stem cells that have become senescent. See, e.g., Park et al., J. Clin. Invest.113:175-79 (2004) and Sousa- Victor, Nature 506:316-21 (2014) describing importance of preventing senescence in stem cells to maintain regenerative capacity of tissues.
  • the effects of the methods of treatment described herein can be analyzed using techniques known in the art, such as comparing symptoms of patients suffering from or at risk of a particular disease or disorder that have received the pharmaceutical composition comprising a compound selected from the table 1 ab and Table 1c with those of patients who were not treated with the compound selected from the table 1 ab and Table 1c or who received a placebo treatment.
  • the terms, “treat” and “treatment,” refer to medical management of a disease, disorder, or condition of a subject (i.e., patient) (see, e.g., Stedman's Medical Dictionary).
  • an appropriate dose and treatment regimen provide the compound selected from the table 1 ab and Table 1c in an amount sufficient to provide therapeutic and/or prophylactic benefit.
  • Therapeutic benefit for subjects to whom the Compounds selected from the table 1 ab and Table 1c described herein are administered includes, for example, an improved clinical outcome, wherein the object is to prevent or slow or retard (lessen) an undesired physiological change associated with the disease, or to prevent or slow or retard (lessen) the expansion or severity of such disease.
  • effectiveness of the one or more Compounds selected from the table 1 ab and Table 1c may include beneficial or desired clinical results that comprise, but are not limited to, abatement, lessening, or alleviation of symptoms that result from or are associated with the disease to be treated; decreased occurrence of symptoms; improved quality of life; longer disease-free status (i.e., decreasing the likelihood or the propensity that a subject will present symptoms on the basis of which a diagnosis of a disease is made); diminishment of extent of disease; stabilized (i.e., not worsening) state of disease; delay or slowing of disease progression; amelioration or palliation of the disease state; and remission (whether partial or total), whether detectable or undetectable; and/or overall survival.
  • beneficial or desired clinical results comprise, but are not limited to, abatement, lessening, or alleviation of symptoms that result from or are associated with the disease to be treated; decreased occurrence of symptoms; improved quality of life; longer disease-free status (i.e., decreasing the likelihood or the propensity
  • the effectiveness of the Compounds selected from the table 1 ab and Table 1c described herein may also mean prolonging survival when compared to expected survival if a subject were not receiving the compound selected from the table 1 ab and Table 1c that selectively kills senescent cells.
  • Administration of a compound selected from the table 1 ab and Table 1c described herein can prolong prolonging survival when compared to expected survival if a subject were not receiving treatment.
  • Subjects in need of treatment include those who already have the disease or disorder as well as subjects prone to have or at risk of developing the disease or disorder, and those in which the disease, condition, or disorder is to be treated prophylactically.
  • a subject may have a genetic predisposition for developing a disease or disorder that would benefit from clearance of senescent cells or may be of a certain age wherein receiving a compound selected from the table 1 ab and Table 1c would provide clinical benefit to delay development or reduce severity of a disease, including an age-related disease or disorder.
  • a compound selected from the table 1 ab and Table 1c would provide clinical benefit to delay development or reduce severity of a disease, including an age-related disease or disorder.
  • the present invention also relates to the following items ITEMS: 1.
  • the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c for ameliorating at list one symptom of the disorder selected from the group, consisting of aging, frailty, senescence, aging related disease, aging related condition, senescence related disease. 3.
  • An anti-aging pharmaceutical composition comprising a compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c . 4.
  • a pharmaceutical composition provided as part of an anti-aging treatment or for treating or preventing an age-related disease or disorder comprising an agent configured to bind to, inhibit, or degrade a protein selected from the group consisting of CDK7 , CHEK2 , CAPN1 , CTSB , HSP90AB1 , HSP90AA1 , HSPA5 , NFKB1 , NPC1 , PABPC1 , ABCB11 , ABCB1 , PLK1 , PLK3 , ADRA2A , ADRA2C , ADRA2B , TUBB6 , SENP8 , SENP6 , SENP7 , MMP14 , MMP13 , MMP8 , MMP3 , MMP1 , MAPKAPK2 , CTSL , CTSK , CTSS , ATXN2 , TBX
  • a pharmaceutical composition provided as part of an anti-aging treatment or for treating or preventing an age-related disease or disorder comprising a compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c and at least one pharmaceutically acceptable excipient.
  • an anti-aging pharmaceutical composition, comprising a human cell modified by the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c . 7.
  • a method of providing an anti-aging treatment or of treating or preventing an age-related disease or disorder of a subject comprising reducing, inhibiting, or degrading a gene or protein selected from the group consisting of: CDK7 , CHEK2 , CAPN1 , CTSB , HSP90AB1 , HSP90AA1 , HSPA5 , NFKB1 , NPC1 , PABPC1 , ABCB11 , ABCB1 , PLK1 , PLK3 , ADRA2A , ADRA2C , ADRA2B , TUBB6 , SENP8 , SENP6 , SENP7 , MMP14 , MMP13 , MMP8 , MMP3 , MMP1 , MAPKAPK2 , CTSL , CTSK , CTSS , ATXN2 , TBXAS1 , CREBBP , PDGFRA , FLT4 , FLT1 , KDR , PDGFRB
  • any one of preceding items further comprising administering to the subject a gene therapy.
  • the age-related disease or disorder is associated with an alleviated level of a protein selected from the group consisting of: CDK7 , CHEK2 , CAPN1 , CTSB , HSP90AB1 , HSP90AA1 , HSPA5 , NFKB1 , NPC1 , PABPC1 , ABCB11 , ABCB1 , PLK1 , PLK3 , ADRA2A , ADRA2C , ADRA2B , TUBB6 , SENP8 , SENP6 , SENP7 , MMP14 , MMP13 , MMP8 , MMP3 , MMP1 , MAPKAPK2 , CTSL , CTSK , CTSS , ATXN2 , TBXAS1 , CREBBP , PDGFRA , FLT4 , FL
  • the age-related disease or disorder is selected from the group consisting of frailty, Alzheimer’s disease, Parkinson’s disease, Huntington’s diseases, cardiovascular disease, renal failure, muscle wasting or cachexia, osteopenia or osteoporosis, obesity, insulin resistance or diabetes, diverse adult-onset cancers, atherosclerosis, cardiovascular disease, adult cancer, arthritis, cataracts, osteoporosis, type 2 diabetes, hypertension, age-progressive dementia; amyotrophic lateral sclerosis, stroke, atrophic gastritis, osteoarthritis, NASH, camptocormia, chronic obstructive pulmonary disease, coronary artery disease, dopamine dysregulation syndrome, metabolic syndrome, effort incontinence, Hashimoto's thyroiditis, heart failure, late life depression, immunosenescence, age related decline in immune response to vaccines, age related decline in response to immunotherapy, myocardial infarction, acute coronary syndrome, sarcopenia, sarcopen
  • the anti-aging treatment is selected from the group consisting of a treatment leading to prevention, amelioration or lessening at least one effect of aging; prevention, amelioration or lessening at least one symptom of aging, prevention, amelioration or lessening at least one symptom of age related disease or condition, decreasing or delaying an increase in a biological age of the subject; slowing a rate of aging of the subject; prevention, amelioration or lessening the effects of frailty; prevention, amelioration or lessening the effects of at least one of an aging-related disease or conditions; increasing a health span or lifespan of the subject; increasing a stress resistance or resilience of the subject; increasing a rate or other enhancement of recovery after surgery, radiotherapy, disease and/or any other stress; prevention, amelioration or lesion the effects of menopausal syndrome; restoring reproductive function; elimination or lessening the spread of senescent cells; modulation of at least one biomarker of aging into the healthier state; a decrease in a
  • the anti-aging treatment is a treatment leading to changing to a healthier state a parameter selected from the group consisting of a a blood parameter, a heart rate, a cognitive function, a bone density, a basal metabolic rate, a systolic blood pressure, a heel bone mineral density (BMD), a heel quantitative ultrasound index (QUI), a heel broadband ultrasound attenuation, a forced expiratory volume in 1-second (FEV1), forced vital capacity (FVC), a peak expiratory flow (PEF), a duration to first press of snap-button in each round, a reaction time, a mean time to correctly identify matches, a right or left hand grip strength, a whole body fat-free mass, a leg fat- free mass, a time for recovery after a stress-inducing event, a resistance to radiation, a morbidity risk, and a mortality risk of the subject.
  • a parameter selected from the group consisting of a a blood parameter, a heart rate, a cognitive function, a bone density,
  • a method of providing an anti-aging treatment or of treating or preventing an age-related disease or disorder of a subject comprising administering to the subject a pharmaceutical composition comprising an inhibitor of a protein selected from the group consisting of: CDK7 , CHEK2 , CAPN1 , CTSB , HSP90AB1 , HSP90AA1 , HSPA5 , NFKB1 , NPC1 , PABPC1 , ABCB11 , ABCB1 , PLK1 , PLK3 , ADRA2A , ADRA2C , ADRA2B , TUBB6 , SENP8 , SENP6 , SENP7 , MMP14 , MMP13 , MMP8 , MMP3 , MMP1 , MAPKAPK2 , CTSL , CTSK , CTSS , ATXN2 , TBXAS1 , CREBBP , PDGFRA , FLT4 , FLT1 , KDR ,
  • An anti-aging pharmaceutical composition comprising a compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c and an enhancer moiety.
  • a kit comprising: a pharmaceutical composition comprising: an agent configured to bind to, inhibit, or degrade a protein selected from the group consisting of: Targets, wherein the agent is selected from the group consisting of a protein, a polymer, an aptamer, a SOMAmer, a peptide, a virus, a small molecule, a nanoparticle, an antibody, a monoclonal antibody, a polyclonal antibody, a humanized monoclonal antibody, a human monoclonal antibody, a human or humanized polyclonal antibody, and at least one pharmaceutically acceptable excipient; and an instruction for using the pharmaceutical composition as part of the anti-aging treatment or to treat or prevent an age-related disease or disorder. 18.
  • a pharmaceutical composition comprising: an agent configured to bind to, inhibit, or degrade a protein selected from the group consisting of: Targets, wherein the agent is selected from the group consisting of a protein, a polymer, an aptamer, a SOMAmer,
  • aging, frailty, an age-related disease or condition is selected from the group consisting of age-related tissue decline, age- related organ decline, degenerative disease, function-decreasing disorder, Alzheimer's disease, Parkinson's disease, cataracts, macular degeneration, glaucoma, atherosclerosis, acute coronary syndrome, myocardial infarction, stroke, recovery after stroke, PSD-95 decrease, hypertension, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), osteoarthritis, type 1 diabetes, obesity, fat dysfunction, coronary artery disease, cerebrovascular disease, periodontal disease, cancer treatment- related disability, chemotherapy treatment-related disability, chemotherapy treatment
  • a method for treating a senescence associated disease or disorder in a subject comprising the subject a the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or combination, comprising such compound, ; wherein the senescence associated disease or disorder is not a cancer, and wherein the such compound or combination is administered during a treatment course of 1-7 days every 0.5-12 months; provided that if the senescence associated disease or disorder is a senescence associated metabolic disorder, the senolytic combination is administered during a treatment course of 1-7 days every 4-12 months.
  • 35. The method, use, compound, composition or kit of any one of preceding items, wherein the pulmonary disease or disorder is idiopathic pulmonary fibrosis or chronic obstructive pulmonary disease.
  • 36. The method, use, compound, composition or kit of any one of preceding items, wherein the neurological disease or disorder is selected from mild cognitive impairment; motor neuron dysfunction; Alzheimer's disease; Parkinson's disease; and macular degeneration.
  • the senescence-associated disease or disorder is a dermatological disease or disorder is selected from eczema, psoriasis, hyperpigmentation, nevi, rashes, atopic dermatitis, urticaria, diseases and disorders related to photosensitivity or photoaging, rhytides; pruritis; dysesthesia; eczematous eruptions; eosinophilic dermatosis; reactive neutrophilic dermatosis; pemphigus; pemphigoid; immunobullous dermatosis; fibrohistocytic proliferations of skin; cutaneous lymphomas; and cutaneous lupus.
  • the method, use, compound, composition or kit of any one of preceding items comprising administering to the subject the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or combination, comprising such compound, , wherein the senolytic combination is administered during a treatment course of 1-7 days every 4-12 months, and wherein the metabolic disease or disorder is selected from diabetes, metabolic syndrome, and obesity. 40.
  • a method for treating a senescence-associated metabolic disease or disorder in a subject comprising administering to the subject the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or combination, comprising such compound, , wherein the senolytic combination is administered during a treatment course of 1-7 days every 4-12 months, and wherein the metabolic disease or disorder is selected from diabetes, metabolic syndrome, and obesity.
  • the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or combination comprising such compound, wherein the senolytic combination is administered during a treatment course of 1-7 days every 4-12 months, and wherein the metabolic disease or disorder is selected from diabetes, metabolic syndrome, and obesity.
  • senescent cell is selected from a senescent fibroblast, a senescent pre-adipocyte, a senescent epithelial cell, a senescent chondrocyte, a senescent neuron, and a senescent endothelial cell. 43. The method, use, compound, composition or kit of any one of preceding items, wherein the senescent cell is a senescent pre-adipocyte 44.
  • the method, use, compound, composition or kit of any one of preceding items comprising administering in or around an eye of the subject a pharmaceutical composition that contains an effective amount of a compound selected from the table 1 ab and Table 1c or a salt thereof.
  • a method of treating an ophthalmic disease or disorder that is not a cancer in a subject the method comprising administering in or around an eye of the subject a pharmaceutical composition that contains an effective amount of a compound selected from the table 1 ab and Table 1c or a salt thereof.
  • the method of any one of preceding items which is a method of treating age-related macular degeneration (AMD).
  • AMD age-related macular degeneration
  • the method of any one of preceding items which is a method of treating glaucoma. 48.
  • the method of any one of preceding items which is a method for preventing or delaying vision loss.
  • 49. The method, use, compound, composition or kit of any one of preceding items, wherein the such method, use, composition or kit is for for preventing or delaying vision loss.
  • 50. The method, use, compound, composition or kit of any one of preceding items, wherein the composition is administered to the eye by intraocular injection.
  • 51. The method, use, compound, composition or kit of any one of preceding items, wherein the composition is administered to the eye by intravitreal injection. 52.
  • the method, use, compound, composition or kit of any one of preceding items whereby p16 positive senescent cells in the eye that are causing one or more symptoms of the ophthalmic disease or disorder are removed from the eye.
  • 53. The method, use, compound, composition or kit of any one of preceding items, wherein the pharmaceutical composition delays disorganization of type IV collagen in the eye.
  • 54. The method, use, compound, composition or kit of any one of preceding items, wherein the pharmaceutical composition is administered in a therapeutically effective course of therapy that includes a period of treatment followed by a non-treatment interval of at least two weeks. 55.
  • a method of treating an ophthalmic disease or disorder in a subject who does not have cancer comprising administering intraocularly into an eye of the subject that is affected with the disease or disorder an effective amount of the compound selected from the table 1 ab and Table 1c , or a pharmaceutically acceptable salt thereof 57.
  • the method, use, compound, composition or kit of any one of preceding items whereby one or more symptoms of an ophthalmic disease or disorder in the eye are decreased in severity or delayed in progression consequent to treating the eye to remove senescent cells.
  • the method, use, compound, composition or kit of any one of preceding items further comprising determining whether positive senescent cells have been removed from the eye of the subject in which senescent cells have been contacted with said benzenesulfonamide 61.
  • the method, use, compound, composition or kit of any one of preceding items further comprising examining the subject to determine whether symptoms of an ophthalmic disease or disorder are decreased in severity or delayed in progression consequent to treating the eye to remove senescent cells. 62.
  • a pharmaceutical composition comprising the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or their pharmaceutically acceptable salt or any combination of it or compounds or , optionally, having the similar SAR characteristics or any it’s pharmaceutically acceptable salt, any it’s hydrate, solvate, tautomer, geometric, optical and stereoisomer thereof, structurally related molecule, structural analog, functional analog, derivative, prodrug, or mixtures thereof in all ratios in therapeutically effective amount and at least one pharmaceutically acceptable excipient for anti-aging use.
  • the method, use, compound, composition or kit of any one of preceding items comprising from 10 mg to 100 mg of compound selected from the table 1 ab and Table 1c as an active ingredient.
  • the method, use, compound, composition or kit of any one of preceding items comprising from 1 mg to 50 mg of active ingredient. 65. The method, use, compound, composition or kit of any one of preceding items, comprising from 1 mg to 75 mg of active ingredient. 66. The method, use, compound, composition or kit of any one of preceding items, comprising from 1 mg to 100 mg of active ingredient. 67. The method, use, compound, composition or kit of any one of preceding items, comprising from 1 mg to 100 mg of active ingredient. 68. The method, use, compound, composition or kit of any one of preceding items, comprising from 1 mg to 25 mg of active ingredient. 69.
  • the method, use, compound, composition or kit of any one of preceding items comprising from 1 mg to 10 mg of active ingredient. 70. The method, use, compound, composition or kit of any one of preceding items, comprising from 1 mg to 5 mg of active ingredient. 71.
  • compositions, pharmaceutical composition or kit of any one of preceding items comprising from 1 mg to 5 g of active ingredient, wherein active ingredient is the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or its pharmaceutically acceptable salt, any it’s hydrate, solvate, tautomer, geometric, optical and stereoisomer thereof, structurally related molecule, structural analog, functional analog, derivative, prodrug, or mixtures thereof in all ratios in therapeutically effective amount and at least one pharmaceutically acceptable excipient.
  • active ingredient is the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or its pharmaceutically acceptable salt, any it’s hydrate, solvate, tautomer, geometric, optical and stereoisomer thereof, structurally related molecule, structural analog, functional analog, derivative, prodrug, or mixtures thereof in all ratios in therapeutically effective amount and at least one pharmaceutically acceptable excipient.
  • 72. The method, use, compound, composition or kit of any one of preceding items, comprising from 1 mg
  • the method, use, compound, composition or kit of any one of preceding items comprising from 10 mg to 75 mg of active ingredient. 74. The method, use, compound, composition or kit of any one of preceding items, comprising from 10 mg to 50 mg of active ingredient. 75. The method, use, compound, composition or kit of any one of preceding items, comprising from 10 mg to 25 mg of active ingredient. 76. The method, use, compound, composition or kit of any one of preceding items, comprising from 1 mg to 10 mg of active ingredient. 77. The method, use, compound, composition or kit of any one of preceding items, wherein composition is for oral administration. 78. The method, use, compound, composition or kit of any one of preceding items, wherein composition is for rectal administration. 79.
  • composition for injection.
  • 80. A use of pharmaceutical composition of any one of the preceding items for anti-aging treatment.
  • 81. The method, use, compound, composition or kit of any one of preceding items, wherein pharmaceutical composition is for anti-aging treatment.
  • 82. A use of the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or any of their pharmaceutically acceptable salts, hydrates, solvates, tautomers, geometric, optical and stereoisomers thereof, structurally related molecules, structural analogs, functional analogs, derivatives, prodrugs, or mixtures thereof in all ratios in therapeutically effective amount for anti-aging treatment.
  • Kit comprising pharmaceutical composition of any one of the preceding items and instruction for using it as an anti-aging treatment.
  • the method, use, compound, composition or kit of any one of preceding items comprising the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or any of their pharmaceutically acceptable salts, hydrates, solvates, tautomers, geometric, optical and stereoisomers thereof, structurally related molecules, structural analogs, functional analogs, derivatives, prodrugs, or mixtures thereof in all ratios in therapeutically effective amount and instruction for its use in anti-aging treatment.
  • Method of anti-aging treatment comprising administering to subject a therapeutically effective amount of the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or any of their pharmaceutically acceptable salts, hydrates, solvates, tautomers, geometric, optical and stereoisomers thereof, structurally related molecules, structural analogs, functional analogs, derivatives, prodrugs, or mixtures thereof in all ratios in therapeutically effective amount or any combination of it.
  • a method of selectively killing one or more senescent cells in a subject in need thereof comprising administering to the subject a composition comprising the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c .
  • the method, use, compound, composition or kit of any one of preceding items further comprising selectively killing one or more senescent cells in a subject in need thereof by the compound selected from the table 1 ab and Table 1c .
  • 97. A method for delaying at least one feature or symptom of aging in a subject, the methodcomprising administering a composition comprising a therapeutically effective amount of the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c . 98.
  • a method for anti-aging treatment comprising administering a composition comprising a therapeutically effective amount of the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c .
  • 99. A method for delaying at least one feature of aging in a subject, the method comprising administering a composition comprising a therapeutically effective amount of the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c .
  • 100. The method, use, compound, composition or kit of any one of preceding items, wherein such method, use, compound, composition or kit for delaying at least one feature of aging in a subject.
  • 101. The method, use, compound, composition or kit of any one of preceding items, wherein the subject has a received DNA- damaging therapy. 102.
  • a pharmaceutical composition of slow or controlled release providing a dosage according any one of preceding items.
  • a drug delivery device providing dosage according any one of preceding items.
  • a dosage of compound selected from the table 1 ab and Table 1c is ranging from about 0.05 ⁇ g/kg to about 100 mg/kg of a patient’s body weight, or 0.01 to about 1000 mg/kg of total body weight per day, or from about 0.1 to about 100 mg/kg of total body weight per day, or from about 0.5 to about 15 mg/kg of total body weight per day, or from about 1 mg/kg to about 50 mg/kg.
  • the method, use, compound, composition or kit of any one of preceding items is for treatment of the disease, condition or disorder selected from the group, consisting of all diseases, conditions and disorders mentioned in this application. 113.
  • compositions or kit of any one of preceding items, wherein treatment comprises reducing, inhibiting, or degrading a protein selected from the group consisting of Targets.
  • An anti-aging pharmaceutical composition comprising a human cell, modified by the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c .
  • a human cell modified by the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c .
  • a pharmaceutical composition comprising a human cell, modified by the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c , wherein such composition is for treatment of disease, disorder or condition selected from this application.
  • a pharmaceutical composition comprising a human cell, modified by the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c is used.
  • the non-limiting example of such cell is hepatocyte, modified by MIZOLASTINE.
  • Another non-limiting example of such cell is thrombocyte , modified by RUPATADINE.
  • Yet another non-limiting example of such cell is brain neuron modified by TRIPROLIDINE.
  • Yet another non-limiting example of such cell is Adipocyte, modified by RUPATADINE.
  • the compound selected from the table 1 ab and Table 1c is in therapeutically effective amount. 124.
  • a pharmaceutical composition, comprising a human senescent cell, modified by the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c is used.
  • a pharmaceutical composition, comprising a human senescent cell, modified by the compound selected from the group consisting of all compounds listed in the Table 1 ab and Table 1c is used.
  • a human senescent cell modified by compound selected from the table 1 ab and Table 1c is used.
  • the non-limiting example of such cell is senescent cell, modified by MIZOLASTINE.
  • Another non-limiting example of such cell is senescent cell , modified by RUPATADINE.
  • Such cell is senescent cell modified by TRIPROLIDINE.
  • Yet another non-limiting example of such cell is senescent cell, modified by any one of the compound selected from the table 1 ab and Table 1c .
  • the compound selected from the table 1 ab and Table 1c is in therapeutically effective amount.
  • the compounds selected from the table 1 ab and Table 1c can be synthesised by the methods known in the art. Some of the compounds selected from the table 1 ab and Table 1c which chemical names are explicitly indicated can be readily obtained from commercial providers.
  • Figure 3 shows the effect in mice of the experimental treatment with the compound selected from Table 1 ab and Table 1c on the Frailty Index (12 and 14 weeks after the intervention).
  • “Antibody” is a small molecule compound selected from the table 1 ab and Table 1c .
  • Figure 4 shows the effects of the experimental treatment in mice with the compound selected from Table 1 ab and Table 1c on the Open Field test results (at the baseline, 4 and 7 weeks after the intervention).
  • “AB3” is a compound selected from the table 1 ab and Table 1c .
  • Figure 5 shows the effects of the experimental treatments in mice with the compound selected from Table 1 ab and Table 1c on the markers of senescence in peripheral lymphocytes (16 and 18 weeks after the intervention).
  • “AB3” is a compound selected from the table 1 ab and Table 1c .
  • Figure 6 shows the effects of the experimental treatments with the compound selected from Table 1 ab and Table 1c on synaptoplasticity in mice (16 and 18 weeks after the intervention).
  • “AB3” is a compound selected from the table 1 ab and Table 1c .
  • Figure 7 [00178]
  • Figure 8 [00179]
  • Figure 14 shows the correlation of dFI (dynamic frailty index) with age. dFI is defined in Avchaciov, K et al. (2020).
  • Figure 15 shows that dFI is associated with the number of senescent cells.
  • a cell includes a single cell as well as a plurality or population of cells.
  • Terms of degree such as “about”, “substantially”, and “approximately” as used herein mean a reasonable amount of deviation of the modified term such that the end result is not significantly changed. These terms of degree should be construed as including a deviation of at least ⁇ 5% of the modified term if this deviation would not negate the meaning of the word it modifies.
  • the terms “subject” and “animal” include all members of the animal kingdom. More specifically, the subject can be a vertebrate, e.g., a mammal such as a mouse, a primate, a simian or a human.
  • Animals include, but are not limited to, farm animals, sport animals, and pets.
  • a subject can be a healthy individual, an individual that has or is suspected of having a disease or a predisposition to the disease, or an individual that is in need of therapy or suspected of needing therapy, or an aged or frail individual.
  • the subject is a mammal.
  • the subject is a human being.
  • the term "a cell" includes a single cell as well as a plurality or population of cells. Cells contemplated with the present application include microbial cells such as bacterial or yeast cells and mammalian cells.
  • amino acid refers to a compound having the following chemical structure: wherein “R” is known as the amino acid residue, or chemical functional group, that is present in the specified amino acid.
  • natural amino acid refers to an amino acid wherein R is one of the chemical functional groups present in amino acids occurring naturally in biological systems.
  • the compound selected from Table 1 ab and Table 1c is useful for treatment or prevention of aging, frailty or an age-related disease or condition.
  • the compound selected from Table 1 ab and Table 1c is modified for cell permeability, improved stability, and/or better bioavailability.
  • the compound selected from Table 1 ab and Table 1c is also or additionally modified with an enhancer moiety.
  • another aspect provides a compound comprising a compound selected from Table 1 ab and Table 1c and an enhancer moiety.
  • the compound selected from Table 1 ab and Table 1c is conjugated directly or indirectly to the enhancer moiety.
  • an enhancer moiety can increase or enhance the activity of the engineered compound.
  • the enhancer may be a permeability enhancer, a stability enhancer and/or a bioavailability enhancer.
  • the enhancer moiety is selected from a protein carrier, or a polymer carrier. In an embodiment, the enhancer moiety is a carrier protein, thereby forming a fusion protein. In another embodiment, the enhancer moiety is a PEG moiety. [00193] In an embodiment, the compound selected from Table 1 ab and Table 1c is conjugated to a carrier protein, thereby forming a fusion protein. [00194] In an embodiment, the compound selected from Table 1 ab and Table 1c of the application has at least one asymmetric center. Where compounds possess more than one asymmetric center, they may exist as diastereomers. It is to be understood that all such isomers and mixtures thereof in any proportion are encompassed within the scope of the present application.
  • stereochemistry of the compounds may be as specified in any given compound selected from Table 1 ab and Table 1c , such compounds, in an embodiment, may also contain certain amounts (for example, less than 20%, suitably less than 10%, more suitably less than 5%) of compounds of the present application having an alternate stereochemistry. It is intended that any optical isomers, as separated, pure or partially purified optical isomers or racemic mixtures thereof are included within the scope of the present application.
  • the compound selected from Table 1 ab and Table 1c and other compounds of the application are pharmaceutically acceptable salts.
  • the pharmaceutically acceptable salt is an acid addition salt or a base addition salt.
  • a suitable salt may be made by a person skilled in the art (see, for example, S. M. Berge, et aI., "Pharmaceutical Salts," J. Pharm. Sci.1977, 66, 1-19).
  • pharmaceutically acceptable means suitable for, or compatible with, the treatment of subjects.
  • An acid addition salt suitable for, or compatible with, the treatment of subjects is any non- toxic organic or inorganic acid addition salt.
  • Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulfuric, nitric and phosphoric acids, as well as acidic metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate.
  • organic acids which form suitable salts include mono-, di- and tricarboxylic acids.
  • organic acids are, for example, acetic, trifluoroacetic, propionic, glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, benzoic, hydroxybenzoic, phenylacetic, cinnamic, mandelic, salicylic, 2-phenoxybenzoic, p-toluenesulfonic acid and other sulfonic acids such as methanesulfonic acid, ethanesulfonic acid and 2-hydroxyethanesulfonic acid.
  • the mono- or di-acid salts are formed, and such salts exist in either a hydrated, solvated or substantially anhydrous form.
  • acid addition salts are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms.
  • the selection criteria for the appropriate salt will be known to one skilled in the art.
  • Other non-pharmaceutically acceptable salts such as but not limited to oxalates may be used, for example in the isolation of compounds of the application for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
  • a base addition salt suitable for, or compatible with, the treatment of subjects is any non- toxic organic or inorganic base addition salt of any acidic compound.
  • Acidic compounds that form a basic addition salt include, for example, compounds comprising a carboxylic acid group.
  • Illustrative inorganic bases which form suitable salts include lithium, sodium, potassium, calcium, magnesium or barium hydroxide as well as ammonia.
  • Illustrative organic bases which form suitable salts include aliphatic, alicyclic or aromatic organic amines such as isopropylamine, methylamine, trimethylamine, picoline, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2- diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like.
  • Exemplary organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
  • the selection of the appropriate salt may be useful, for example, so that an ester functionality, if any, elsewhere in a compound is not hydrolyzed.
  • the selection criteria for the appropriate salt will be known to one skilled in the art.
  • Salts of the compounds of the application are generally formed by dissolving the neutral compound in an inert organic solvent and adding either the desired acid or base and isolating the resulting salt by either filtration or other known means.
  • Solvates of compounds of the application include, for example, those made with solvents that are pharmaceutically acceptable.
  • solvates examples include water (resulting solvate is called a hydrate) and ethanol and the like.
  • solvates are formed by dissolving the compound in the appropriate solvent and isolating the solvate by cooling or using an antisolvent.
  • the solvate is typically dried or azeotroped under ambient conditions. The selection of suitable conditions to form a particular solvate can be made by a person skilled in the art.
  • the compounds of the present application further exist in varying polymorphic forms and it is contemplated that any polymorphs, or mixtures thereof, are included within the scope of the present application
  • Compositions [00201]
  • the application also includes a composition, optionally a pharmaceutical composition, comprising the compound selected from Table 1 ab and Table 1c or other compounds described herein.
  • the composition comprises a carrier or diluent.
  • the carrier is a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
  • Suitable carriers are described in the most recent edition of Remington’s Pharmaceutical Sciences, a standard reference text in the field, which is incorporated herein by reference.
  • Optional examples of such carriers or diluents include, but are not limited to, water, saline, ringer’s solutions, dextrose solution, and 5% human serum albumin and bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • routes of administration include parenteral (e.g., intravenous, subcutaneous, intramuscular), intradermal, intraperitoneal, subcutaneous, intranasal, epidural, sublingual, intravaginal, rectal, by inhalation, topical intracerebral, oral, intranasal, buccal, rectal, or transdermal administration routes.
  • parenteral e.g., intravenous, subcutaneous, intramuscular
  • intradermal intraperitoneal
  • subcutaneous, intranasal, epidural sublingual
  • intravaginal intravaginal
  • rectal by inhalation
  • topical intracerebral e.g., topical intracerebral
  • oral, intranasal, buccal, rectal, or transdermal administration routes e.g., transdermal administration routes.
  • the composition described herein is formulated for local administration.
  • composition described herein may introduced into the central nervous system, circulatory system or gastrointestinal tract by any suitable route, including intraventricular, intrathecal injection, paraspinal injection, epidural injection, enema, and by injection adjacent to a peripheral nerve.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent, or via perfusion in a fluorocarbon or synthetic pulmonary surfactant.
  • oral or parenteral compositions are formulated in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the carrier is selected on the basis of compatibility with the compound disclosed herein, and the release profile properties of the desired dosage form.
  • exemplary carriers include, e.g., binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, and the like.
  • Pharmaceutically compatible carrier materials include, but are not limited to, acacia, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, maltodextrin, glycerine, magnesium silicate, polyvinylpyrrollidone (PVP), cholesterol, cholesterol esters, sodium caseinate, soy lecithin, taurocholic acid, phosphotidylcholine, sodium chloride, tricalcium phosphate, dipotassium phosphate, cellulose and cellulose conjugates, sugars sodium stearoyl lactylate, carrageenan, monoglyceride, diglyceride, pregelatinized starch, and the like.
  • PVP polyvinylpyrrollidone
  • the carrier protects the compound against rapid elimination from the body, such as in a sustained/controlled release formulation, including implants and microencapsulated delivery systems.
  • compositions further include pH adjusting agents or buffering agents which include acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids, bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane, and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids
  • bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane
  • buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • compositions include one or more salts in an amount required to bring osmolality of the composition into an acceptable range.
  • salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions
  • suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
  • the compositions include, but are not limited to, sugars like trehalose, sucrose, mannitol, maltose, glucose, or salts like potassium phosphate, sodium citrate, ammonium sulfate and/or other agents such as heparin to increase the solubility and in vivo stability of polypeptides.
  • the compositions further include diluents which are used to stabilize compounds because they can provide a more stable environment. Salts dissolved in buffered solutions (which also can provide pH control or maintenance) are utilized as diluents in the art, including, but not limited to a phosphate buffered saline solution.
  • diluents increase bulk of the composition to facilitate compression or create sufficient bulk for homogenous blend for capsule filling.
  • Such compounds can include e.g., lactose, starch, mannitol, sorbitol, dextrose, microcrystalline cellulose such as Avicel®, dibasic calcium phosphate, dicalcium phosphate dihydrate, tricalcium phosphate, calcium phosphate, anhydrous lactose, spray-dried lactose, pregelatinized starch, compressible sugar, such as Di- Pac® (Amstar), mannitol, hydroxypropylmethylcellulose, hydroxypropylmethylcellulose acetate stearate, sucrose-based diluents, confectioner’s sugar, monobasic calcium sulfate monohydrate, calcium sulfate dihydrate, calcium lactate trihydrate, dextrates, hydrolyzed cereal solids, amylose, powdered cellulose, calcium carbonate, glycine, kaolin, manni
  • compositions include disintegration agents or disintegrants to facilitate the breakup or disintegration of a substance.
  • disintegrate include both the dissolution and dispersion of the dosage form when contacted with gastrointestinal fluid.
  • disintegration agents include a starch, e.g., a natural starch such as corn starch or potato starch, a pregelatinized starch such as National 1551 or Amijel®, or sodium starch glycolate such as Promogel® or Explotab®, a cellulose such as a wood product, methylcrystalline cellulose, e.g., Avicel®, Avicel® PH101, Avicel® PH102, Avicel® PH105, Elcema® P100, Emcocel®, Vivacel®, Ming Tia®, and Solka-Floc®, methylcellulose, croscarmellose, or a cross-linked cellulose, such as cross-linked sodium carboxymethylcellulose (Ac-Di- Sol®), cross-linked carboxymethylcellulose, or cross-linked croscarmellose, a cross-linked starch such as sodium starch glycolate, a cross-linked polymer such as crospovidone, a cross-linked polyvinylpyrrolidone, al
  • the compositions include filling agents such as lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.
  • Lubricants and glidants are also optionally included in the compositions described herein for preventing, reducing or inhibiting adhesion or friction of materials.
  • Exemplary lubricants include, e.g., stearic acid, calcium hydroxide, talc, sodium stearyl fumerate, a hydrocarbon such as mineral oil, or hydrogenated vegetable oil such as hydrogenated soybean oil (Sterotex®), higher fatty acids and their alkali-metal and alkaline earth metal salts, such as aluminum, calcium, magnesium, zinc, stearic acid, sodium stearates, glycerol, talc, waxes, Stearowet®, boric acid, sodium benzoate, sodium acetate, sodium chloride, leucine, a polyethylene glycol (e.g., PEG-4000) or a methoxypolyethylene glycol such as CarbowaxTM, sodium oleate, sodium benzoate, glyceryl behenate, polyethylene glycol, magnesium or sodium lauryl sulfate, colloidal silica such as SyloidTM, Cab-O-Sil®, a starch such as corn starch, silicone
  • Plasticizers include compounds used to soften the microencapsulation material or film coatings to make them less brittle. Suitable plasticizers include, e.g., polyethylene glycols such as PEG 300, PEG 400, PEG 600, PEG 1450, PEG 3350, and PEG 800, stearic acid, propylene glycol, oleic acid, triethyl cellulose and triacetin. Plasticizers can also function as dispersing agents or wetting agents.
  • Solubilizers include compounds such as triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, sodium lauryl sulfate, sodium doccusate, vitamin E TPGS, dimethylacetamide, N- methylpyrrolidone, N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropylmethyl cellulose, hydroxypropyl cyclodextrins, ethanol, n-butanol, isopropyl alcohol, cholesterol, bile salts, polyethylene glycol 200-600, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide and the like.
  • Stabilizers include compounds such as any antioxidation agents, buffers, acids, preservatives and the like.
  • Exemplary stabilizers include L-arginine hydrochloride, tromethamine, albumin (human), citric acid, benzyl alcohol, phenol, disodium biphosphate dehydrate, propylene glycol, metacresol or m-cresol, zinc acetate, polysorbate-20 or Tween® 20, or trometamol.
  • Suspending agents include compounds such as polyvinylpyrrolidone, e.g., polyvinylpyrrolidone K12, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30, vinyl pyrrolidone/vinyl acetate copolymer (S630), polyethylene glycol, e.g., the polyethylene glycol can have a molecular weight of about 300 to about 6000, or about 3350 to about 4000, or about 7000 to about 5400, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, hydroxymethylcellulose acetate stearate, polysorbate-80, hydroxyethylcellulose, sodium alginate, gums, such as, e.g., gum tragacanth and gum acacia, guar gum, xanthans, including xanthan gum, sugars, cellulosics, such as,
  • Surfactants include compounds such as sodium lauryl sulfate, sodium docusate, Tween 60 or 80, triacetin, vitamin E TPGS, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic® (BASF), and the like.
  • compounds such as sodium lauryl sulfate, sodium docusate, Tween 60 or 80, triacetin, vitamin E TPGS, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic® (BASF), and the like.
  • Pluronic® Pluronic®
  • Additional surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil, and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40. Sometimes, surfactants is included to enhance physical stability or for other purposes.
  • Viscosity enhancing agents include, e.g., methyl cellulose, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, hydroxypropylmethyl cellulose acetate stearate, hydroxypropylmethyl cellulose phthalate, carbomer, polyvinyl alcohol, alginates, acacia, chitosans and combinations thereof.
  • Wetting agents include compounds such as oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, sodium docusate, sodium oleate, sodium lauryl sulfate, sodium doccusate, triacetin, Tween 80, vitamin E TPGS, ammonium salts and the like.
  • the compositions include, but are not limited to, aqueous fluid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations (e.g., nanoparticle formulations), and mixed immediate and controlled release formulations.
  • the amount of composition described herein that is effective for treating a corresponding disease or condition can be determined using standard clinical or pharmacokinetic techniques known to those with skill in the art.
  • in vitro or in vivo assays can optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed can also depend on the route of administration, the disease or condition, the seriousness of the disease or condition being treated, as well as various physical factors related to the individual being treated, and can be decided according to the judgment of a health- care practitioner.
  • the composition is a formulation for administration, in an amount of compound selected from the table 1 ab and Table 1c , binding agent, peptide, nucleic acid and/or recombinant cell ranging from about 0.05 ⁇ g/kg to about 100 mg/kg of a patient’s body weight, or 0.01 to about 1000 mg/kg of total body weight per day, or from about 0.1 to about 100 mg/kg of total body weight per day, or from about 0.5 to about 15 mg/kg of total body weight per day, or from about 1 mg/kg to about 50 mg/kg of total body weight.
  • the composition is formulated for administration in one or multiple doses per day or per week or per month or per 6 months or per year or per 3 years or per 8 years or per 12 years or once in a lifetime.
  • the composition is formulated for equivalent dosages which can be administered over various time periods including, but not limited to, about every 2 hours, about every 4 hours, about every 8 hours, about every 12 hours, about every 24 hours, about every 36 hours, about every 48 hours, about every 72 hours, about every week, about every two weeks, about every three weeks, about every month, and about every two months or every 6 months or every year or every 3 years or every 8 years or every 12 years or once in a lifetime or daily lifelong or as decided by practitioner or patient.
  • the number and frequency of dosages corresponding to a completed course of therapy can be determined according to the judgment of a health-care practitioner.
  • the administration is suspended or dosage decreased until the toxicity or adverse effects disappear and then the administration and/or dosage is resumed on the previous level.
  • the dose for anti-aging use or any other use disclosed in this application is the same or about the same as an effective dose of the compound for its initial indication.
  • the dose for anti-aging use or any other use disclosed in this application is the same or about the same as an effective dose of the compound for the indication for which such compound was approved.
  • kits [00228] Also provided herein is a kit, comprising a compound selected from Table 1 ab and Table 1c or composition disclosed in this application and optionally a description or instruction for its use and additionally optionally further comprising medication labeling information.
  • the instruction is an instruction for use of the compound selected from Table 1 ab and Table 1c or composition comprising such compound for treating or preventing aging, frailty or an age-related disease or condition in a subject, for delaying one or more signs or symptoms of aging in a subject and/or for increasing longevity in a subject.
  • the instruction is an instruction for use of the compound selected from Table 1 ab and Table 1c or composition comprising such compound for treating or preventing a senescence associated disease or disorder in a subject and for selectively killing one or more senescent cells in a subject in need thereof.
  • the instruction is an instruction for use of the compound selected from Table 1 ab and Table 1c or composition comprising such compound for treating or preventing viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject or for alleviating signs and symptoms of viral disease, optionally COVID-19 or influenza, or infectious disease in a subject.
  • the application also provides uses and methods relating to the compound selected from Table 1 ab and Table 1c or composition comprising such compound.
  • the activity of the target, selected from the Targets tt can be inhibited by at least 5, 10, 15, 25, 50, 75 or 100%. Inhibiting includes any decrease in activity as compared to activity under otherwise the same conditions except in the absence of the binding agent.
  • the compounds selected from Table 1 ab and Table 1c or compositions comprising such compounds are useful for treating or preventing a condition selected from the group consisting of: aging, frailty, an age-related disease or condition in a subject, for delaying one or more signs or symptoms of aging in a subject and/or for increasing longevity in a subject.
  • the compounds selected from Table 1 ab and Table 1c or compositions comprising such compounds, compounds and compositions of the present application are also useful for treating or preventing a senescence associated disease or disorder in a subject and for selectively killing one or more senescent cells in a subject in need thereof.
  • the compounds selected from Table 1 ab and Table 1c or compositions comprising such compounds are used in a method for treating or preventing aging, frailty, an age-related disease or condition in a subject, for delaying one or more signs or symptoms of aging in a subject, for increasing longevity in a subject, treating or preventing a senescence associated disease or disorder in a subject and/or for selectively killing one or more senescent cells in a subject, the method comprising administering an effective amount of the compound selected from Table 1 ab and Table 1c or composition comprising such compound to a subject in need thereof.
  • a use of a compound selected from Table 1 ab and Table 1c or composition comprising such compound for treating or preventing aging, frailty or an age-related disease or condition in a subject, for reversing or delaying one or more signs or symptoms of aging in a subject, for increasing longevity in a subject, treating or preventing a senescence associated disease or disorder in a subject and/or for selectively killing one or more senescent cells in a subject is included.
  • a use of compound selected from Table 1 ab and Table 1c or composition comprising such compound for preparation of a medicament for treating or preventing aging, frailty or an age-related disease or condition in a subject, for delaying one or more signs or symptoms of aging in a subject, for increasing longevity in a subject, treating or preventing a senescence associated disease or disorder in a subject and/or for selectively killing one or more senescent cells in a subject is provided.
  • a compound selected from Table 1 ab and Table 1c or composition comprising such compound is for use in treating or preventing aging, frailty or an age-related disease or condition in a subject, for delaying one or more signs or symptoms of aging in a subject, for increasing longevity in a subject, treating or preventing a senescence associated disease or disorder in a subject and/or for selectively killing one or more senescent cells in a subject is included.
  • a compound selected from Table 1 ab and Table 1c or composition comprising such compound or other compound or composition disclosed herein is for use in preventing or slowing down the progression of senescence or some of its features.
  • a compound selected from Table 1 ab and Table 1c or composition comprising such compound, compound or composition disclosed herein for interfering with the progression of cells entering senescence or modulating their activity by reducing senescence-associated secretory phenotype (SASP) generation.
  • a compound selected from Table 1 ab and Table 1c or composition comprising such compound, or composition disclosed herein for use as senostatic.
  • the phrase “treating” includes reversing, alleviating or inhibiting the progression of a disease or condition or symptoms or conditions associated the disease or condition. “Treating” also includes extending survival in a subject.
  • the term “aging” refers to an age-dependent or age-progressive decline in intrinsic physiological function.
  • the expression “treating or preventing aging, frailty or an age-related disease or condition” relates to the amelioration at least one symptom of the age-related condition in the subject, amelioration at least one symptom of the age-related disease in the subject, amelioration or lessening of the effects of aging, decreasing or delaying an increase in the biological age, slowing rate of aging; treatment, prevention, amelioration and lessening the effects of frailty or at least one of aging related diseases and conditions or declines or slowing down the progression of such decline (including but not limited to those indicated in Table 4, “Declines”), condition or disease, increasing health span or lifespan, rejuvenation, increasing stress resistance or resilience, increasing rate or other enhancement of recovery after surgery, radiotherapy, disease and/or any other stress, decreasing all-causes or multiple causes of mortality risks or mortality risks
  • the treatment leading to the modulating of at least one of biomarkers of aging into more youthful state or slowing down its change into “elder” state is also regarded to be an anti-aging treatment, including but not limited to biomarkers of aging which are visible signs of aging, such as wrinkles, grey hairs etc.
  • an age-related disease or condition is selected from: age-related tissue decline, age-related organ decline, degenerative disease, function-decreasing disorder, Alzheimer's disease, Parkinson's disease, cataracts, macular degeneration, glaucoma, atherosclerosis, acute coronary syndrome, myocardial infarction, stroke, recovery after stroke, PSD-95 decrease, hypertension, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), osteoarthritis, type 2 diabetes, obesity, fat dysfunction, coronary artery disease, cerebrovascular disease, periodontal disease, cancer treatment-related disability, chemotherapy treatment-related disability, chemotherapy treatment-related frailty, frailty, radiotherapy and other radiation related disability, chemotherapy treatment-related frailty, cancer treatment-related atrophy, cancer treatment-related fibrosis, brain injury, heart injury, and therapy-related myelodysplastic syndrome, accelerated aging, accelerated aging disease, Hutchinson-Gilford pro
  • “treating or preventing aging, frailty or an age-related disease or condition” includes cell tumorigenesis through autophagy-related cell death or autophagy induction.
  • “treating or preventing aging, frailty or an age-related disease or condition” comprises treatment of any one of the disease, condition or decline known in the art as treated by any one of anti- aging interventions, including but not limited to metformin, NAD+, melatonin, rapamycin, any rapalog, any senolytic, BASISTM, caloric restriction or 16/8 intermittent fasting.
  • Aging-related changes in any parameter or physiological metric are also regarded as age- related conditions, including but not limited to aging related change in blood parameters, heart rate, cognitive functions/decline, bone density, basal metabolic rate, systolic blood pressure, heel bone mineral density (BMD), heel quantitative ultrasound index (QUI), heel broadband ultrasound attenuation, heel broadband ultrasound attenuation, forced expiratory volume in 1-second (FEV1), forced vital capacity (FVC), peak expiratory flow (PEF), duration to first press of snap-button in each round, reaction time, mean time to correctly identify matches, hand grip strength (right and/or left), whole body fat-free mass, leg fat-free mass (right and/or left), and time for recovery after any stress (wound, operation, chemotherapy, disease, change in lifestyle etc.).
  • Aging related change in any parameter of organism is also regarded as an aging related condition, including but not limited to aging related change in at least one of the parameter selected from the Table 4 “Declines”.
  • the signs and symptoms of aging in mammals such as humans include, but are not limited to, hearing loss, cognitive decline, wrinkles, fertility decline, hair greying, osteoarthritis, frailty, atherosclerosis, generalized organ atrophy, diminished stress tolerance and reduced longevity. The aging process is also manifested at the cellular level.
  • signs and symptoms of cellular aging include, but are not limited to, loss of doubling capacity, increased levels of apoptosis, changes in differentiated phenotype, and changes in metabolism, e.g., decreased levels of protein synthesis and turnover. Telomere shortening may also be an indicator of aging. In an embodiment, any of the signs and symptoms described above are decreased by at least 5, 10, 25, 50, 75 or 100% compared to what would be expected without treatment as described herein. [00244] The length of time from birth to death is known as the lifespan of an organism, and each organism has a characteristic average lifespan.
  • non-immortal cells or cell lines are characterized by a predictable lifespan in vitro, broadly divisible into three phases corresponding to growth, maturation, and decline (i.e., senescence).
  • the lifespans of many non-immortal cells in culture, particularly mammalian cells frequently varies from only a matter of hours to only several weeks, even under optimal culture conditions. Even “immortal cells” tend to lose viability as a function of time in culture, with corresponding decline of the cell mass.
  • the term “increasing longevity” includes extending lifespan.
  • lifespan is extended by at least 1, 3 or 5 days, 1, 2, 3, 4, 5 or 6 weeks, 1, 2, 3, 6 or 12 months, or at least 2, 3, 4, 5, 10, 15, 20, 25, 30 or 40 years over the lifespan that would be expected without treatment as described herein.
  • an “FSTL3 associated disease or condition” is a disease or condition that has a relationship with the presence, absence, level and/or function of FSTL3 or a disease or condition that is characterized by the presence, absence, level and/or function of FSTL3.
  • Gao et al. (2020) have shown that up-regulation of FSTL3 promotes proliferation and migration of non-small cell lung cancer cells.
  • FSTL3 inhibition in particular embodiments, has one or more activities on pancreatic islet cells selected from the group consisting of increasing insulin secretion from beta cells, increasing beta cell regeneration, promoting transdifferentiation of an alpha cell or any other pancreatic cell to a beta cell, and inhibiting transdifferentiation of a beta cell to an alpha cell, or any combination thereof. It has also been shown that FSTL3 Neutralizing Antibodies Restore Function to Diabetic Mouse and Human Islets (Schneyer et al., 2020). In an embodiment, a compound selected from the table 1 ab and Table 1c or composition, comprising such compound is useful for treatment of FSTL3 associated disease or condition.
  • an age-related disease or condition is cancer, optionally a cancer selected from: non-small cell lung cancer, atypical teratoid rhabdoid tumor, brain tumor, anal cancer, astrocytoma, vaginal cancer, extrahepatic bile duct cancer, intraocular melanoma, hairy cell leukemia, hepatocellular liver cancer, gestational trophoblastic disease , germ cell tumor, hypopharyngeal cancer, histiocytosis, histiocytosis Langerhans, high-grade astrocytoma, astrocytoma, glioma, brain stem glioma, invasive lobular carcinoma, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, cutaneous T- cell lymphoma, peripheral T-cell lymphoma, non-specific lymphoma mantle cell, lymphogranulomatosis, colorectal cancer, craniopharyngioma, le
  • the age-related disease or condition is diabetes, optionally type 1 diabetes or type 2 diabetes.
  • the age-related disease or condition is a cardiovascular disorder, a cognitive disorder, a neurodegenerative disorder, a metabolic disorder, a muscular disorder, cardiac hypertrophy, diastolic heart, left ventricle wall thickness, myocardial fibrosis, or both left ventricle wall thickness and myocardial fibrosis any condition set out in WO2016049662.
  • the age-related disease or condition comprises at least one symptom of diastolic heart failure in a subject in need thereof.
  • the subject has preserved ejection fraction but elevated left ventricular diastolic pressure (LVDP), as compared to subjects without diastolic heart failure.
  • LVDP left ventricular diastolic pressure
  • the subject has preserved ejection fraction but elevated myocardial fibrosis, as compared to subjects without diastolic heart failure.
  • the age-related disease or condition comprises diastolic heart failure.
  • Cellular senescence is characterized by the cessation of cell division.
  • the expression “selectively killing one or more senescent cells refers to "selectively" (preferentially or to a greater degree) destroying, killing, removing, or facilitating selective destruction of senescent cells.
  • the binding agent or composition destroys or kills a senescent cell in a biologically, clinically, and/or statistically significant manner compared with its capability to destroy or kill a non- senescent cell.
  • the binding agent or composition is used in an amount and for a time sufficient that selectively kills established senescent cells but is insufficient to kill (destroy, cause the death of) a non-senescent cell in a clinically significant or biologically significant manner.
  • the compound selected from the table 1 or composition, comprising such compound selectively or differentially or disproportionately kills senescent cells over non-senescent cells when administered systemically or locally to a mammal.
  • senescent cells are killed, for example by apoptosis.
  • “selectively killing one or more senescent cells” is assessed, for example, by the LD50 of the compound selected from the table 1 or composition, comprising such compound, wherein the LD50 of the compound selected from the table 1 or composition, comprising such compound, in non-senescent cells is greater than 3 times than the LD50 of the composition in senescent cells.
  • killing is measured as a reduction in viable cells. In an embodiment, the reduction in viable cells is greater than 15% following the methods and uses described herein.
  • a senescence associated disease or disorder in a subject refers to a disease or disorder which is fully or partially mediated by the induction or maintenance of a non- proliferating or senescent state in a cell or a population of cells in a subject.
  • Non-limiting examples of senescence associated diseases or disorders include cardiovascular diseases such as angina, aortic aneurysm, arrhythmia, brain aneurysm, cardiac diastolic dysfunction, cardiac fibrosis, cardiac stress resistance, cardiomyopathy, carotid artery disease, coronary thrombosis, endocarditis, hypercholesterolemia, hyperlipidemia, mitral valve prolapsed, and peripheral vascular disease; inflammatory or autoimmune diseases such as herniated intervertebral disc, inflammatory bowel disease, kyphosis, oral mucositis, lupus, interstital cystitis, scleroderma, and alopecia; neurodegenerative diseases such as dementia, Huntington's disease, motor neuron dysfunction, age-related memory decline, and depression/mood disorders; metabolic diseases such as diabetic ulcer and metabolic syndrome; pulmonary diseases such as age-related loss of pulmonary function, asthma, bronchiectasis, cystic fibrosis, emphy
  • Mortality from acute respiratory diseases including pneumonia, influenza and COVID-19 is strongly age dependent. Accordingly, a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compounds and compositions of the present application are useful for treating or preventing viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject or for alleviating signs and symptoms of viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject.
  • a compound selected from the table 1 ab and Table 1c or composition comprising such compound, compounds and compositions described herein are used in a method for treating or preventing viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject or for alleviating signs and symptoms of viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject, the method comprising administering an effective amount of a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition disclosed herein to a subject in need thereof.
  • a use of a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition disclosed herein for treating or preventing viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject or for alleviating signs and symptoms of viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject is included.
  • a compound selected from the table 1 ab and Table 1c or composition comprising such compound, other compound or composition disclosed herein for use in treating or preventing viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject or for alleviating signs and symptoms of viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject is included.
  • a use of a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition disclosed herein for preparation of a medicament for treating or preventing viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject or for alleviating signs and symptoms of viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject is provided.
  • a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition disclosed herein is for use in treating or preventing viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject or for alleviating signs and symptoms of viral disease, optionally COVID-19 or influenza, or infectious disease, or pneumonia in a subject.
  • the viral disease is COVID-19, or a disease, caused by SARS-CoV, SARS-CoV-2 (Severe acute respiratory syndrome coronavirus 2) or other coronavirus.
  • a compound selected from the table 1 ab and Table 1c or composition, comprising such compound is used for reduction of pulmonary fibrosis caused by viral infection of SARS-CoV, reduction of pulmonary fibrosis caused by viral infection of SARS-CoV-2, reduction of virus titer of SARS-CoV and/or reduction of virus titer of SARS-CoV-2.
  • a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition and an immunotherapy are used in a method of treating a cancer or of an infectious disease in a subject in need thereof, the method comprising administering an effective amount of a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition and an immunotherapy disclosed herein to a subject in need thereof.
  • any other immunotherapy is used.
  • a use of a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition disclosed herein and an immunotherapy for treating a cancer or an infectious disease is included.
  • a use of an A compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition disclosed herein and an immunotherapy for preparation of a medicament for treating a cancer or an infectious disease is provided.
  • a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition disclosed herein and an immunotherapy is for treating a cancer or an infectious disease.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition disclosed herein is an adjuvant for the immunotherapy.
  • the cancer is a solid cancer selected from the group consisting of melanoma, breast carcinoma, colon carcinoma, renal carcinoma, adrenocortical carcinoma, testicular teratoma, skin sarcoma, fibrosarcoma, lung carcinoma, adenocarcinoma, liver carcinoma, glioblastoma, prostate carcinoma and pancreatic carcinoma or any other cancer described in this application.
  • the infectious disease caused by a virus, a bacterium, a fungus or a protozoan parasite.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition disclosed herein can be used or administered prior to and/or concomitantly with the immunotherapy.
  • said immunotherapy comprises an adoptive transfer of immune cells, for example, T cells, natural killer (NK) cells, CAR T cells, CAR NK cells, autologous immune cells or CD8+ T cells.
  • said immunotherapy comprises a checkpoint inhibitor for example inhibitors of PD-l such as pembrolizumab, nivolumab, cemiplimab, tislelizumab, spartalizumab, ABBV- 181 and JNJ- 63723283, inhibitors of PD-L1 such as avelumab, atezolizumab and durvalumab, inhibitors of CTLA-4 such as ipilimumab and tremelimumab, and any mixtures thereof.
  • PD-l such as pembrolizumab, nivolumab, cemiplimab, tislelizumab, spartalizumab, ABBV- 181 and JNJ- 63723283
  • inhibitors of PD-L1 such as avelumab, atezolizumab and durvalumab
  • inhibitors of CTLA-4 such as ipilimumab and tremelim
  • said immunotherapy comprises ipilimumab, Tremelimumab, AGEN1181, BMS-986218, BMS-986205, REGN4659, SDREGN2810, ADU-1604, Balstilimab, cemiplimab, nivolumab, pembrolizumab, avelumab, durvalumab, atezolizumab, Cemiplimab, BCD-217, BCD-100, BCD-145, Pembrolizumab, Vopratelimab, JTX-2011, MEDI4736, Camrelizumab (SHR-1210), Lirilumab, CS1002, CS1003, Relatlimab, AntiCD137, AK104, XmAb20717, NKTR-214 and any mixtures thereof.
  • said immunotherapy comprises a vaccination.
  • the immunotherapy is selected from the group consisting of: PD- 1 inhibitor, PD-L1 inhibitor, T-Cell stimulant, CTLA antibody, T-cell surface glycoprotein, CD3 antibody, T-cell surface glycoprotein CD3 antibody, rituximab (Rituxan), Brentuximab Vedotin (Adcetriz), Ado- trastuzumab emtansine (Kadcyla) Cetuximab (Erbitux), bevacizumab (Avastin), Ibritumomab (Zevalin), vedolizumab (Entyvio), pembrolizumab (Keytruda), Alemtuzamab atezolizumab (Tecentriq), avelumab (Bavencio), durvalumab (Imfinzi), B-701, Ofatumumab, Obinut
  • the subject is a subject who is an “aged subject”.
  • An “aged subject” is understood as a human being of chronological age (or in some embodiments, of biological age) of 30 years or older, 35 years or older, 40 years or older, 45 years or older, 50 years or older, 55 years or older, 60 years or older, 65 years or older, 70 years or older, 75 years or older, 80 years or older, 85 years or older, 90 years or older, 95 years or older.
  • aged subject is understood as a frail subject.
  • the compounds selected from the table 1 ab and Table 1c or compositions, comprising such compounds, other compounds and compositions described herein are for use parenterally (e.g., intravenously, subcutaneously, intramuscularly), intradermally, intraperitoneally, subcutaneously, intranasally, epidurally, sublingually, intravaginally, rectally, by inhalation, topical intracerebrally, orally, intranasally, buccally, or transdermally or are administered parenterally (e.g., intravenously, subcutaneously, intramuscularly), intradermally, intraperitoneally, subcutaneously, intranasally, epidurally, sublingually, intravaginally, rectally, by inhalation, topical intracerebrally, orally, intranasally, buccally, or transdermally.
  • parenterally e.g., intravenously, subcutaneously, intramuscularly
  • the compounds selected from the table 1 ab and Table 1c or compositions, comprising such compounds are for use topically or are administered topically.
  • An effective amount of a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition of the application relates generally to the amount needed to achieve a desired objective.
  • the amount required to be administered will furthermore depend on the activity of the compounds selected from the table 1 ab and Table 1c or compositions, comprising such compound will also depend on the rate at which an administered compound is depleted from the free volume of the subject to which it is administered.
  • Common ranges for effective dosing of a compound of the application may be, by way of non-limiting example, from about 0.01 mg kg body weight to about 50 mg/kg body weight. Common dosing frequencies may range, for example, from twice daily to once a week.
  • a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition is used in combination with at least one additional agent (for example, an immunotherapy).
  • the additional agent is administered prior to, overlapping with, concurrently, and/or after administration of a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition.
  • a compound selected from the table 1 ab and Table 1c or composition comprising such compound, other compounds or composition and additional agent are administered in a single formulation or in separate formulations, and if administered separately, then optionally, by different modes of administration.
  • the combination of a compound selected from the table 1 ab and Table 1c or composition, comprising such compound, other compound or composition of this disclosure and one or more other agents act synergistically.
  • a non-human animal may refer to one or more non-human animals, or a plurality of such animals
  • reference to "a cell” or “the cell” includes reference to one or more cells and equivalents thereof (e.g., plurality of cells) known to those skilled in the art, and so forth.
  • At least one for example, when referring to at least one compound or to at least one composition, has the same meaning and understanding as the term, "one or more.
  • Disclosed herein are compounds, compositions and combinations, kits, and methods for anti-aging treatment and related mediums and methods.
  • lifespan extension goes together with other anti-aging effects, such as, but not limited to healthspan extension, rejuvenation, prevention and treatment of diverse age-related diseases, disorders and declines (including but not limited to Alzheimer’s, Parkinson’s, and Huntington’s diseases, cardiovascular disease, renal failure, muscle wasting [cachexia], osteopenia or osteoporosis, obesity, insulin resistance or diabetes, and diverse adult-onset cancers).
  • the term “protocol” is used in the meaning – the combination of formulation, dosage, regimen, root of administration used.
  • the reference that the compound is used by the same protocol as it was used in phase 2 clinical trials means that such compound is used in the same formulation, dosage, regimen and root of administration as it was used in such phase 2 clinical trial.
  • the term “Targets” means all the following proteins: CDK7 , CHEK2 , CAPN1 , CTSB , HSP90AB1 , HSP90AA1 , HSPA5 , NFKB1 , NPC1 , PABPC1 , ABCB11 , ABCB1 , PLK1 , PLK3 , ADRA2A , ADRA2C , ADRA2B , TUBB6 , SENP8 , SENP6 , SENP7 , MMP14 , MMP13 , MMP8 , MMP3 , MMP1 , MAPKAPK2 , CTSL , CTSK , CTSS , ATXN2 , TBXAS1 , CREBBP , PDGFRA , FLT4 , FLT1 , KDR , PDGFRB , FLT3 , KIT , CSF1R , IKBKB , IKBKE ,
  • Target means any one protein selected from the group consisting of Targets.
  • the term “subject,” as used herein, generally refers to an animal, such as a mammalian species (e.g., mouse or human) or avian (i.e., bird) species, nematode (e.g., C. elegans), or other organism, such as a plant. More specifically, the subject can be a vertebrate, e.g., a mammal such as a mouse, a primate, a simian or a human. Animals include, but are not limited to, farm animals, sport animals, and pets.
  • a subject can be a healthy individual, an individual that has or is suspected of having a disease or a predisposition to the disease, or an individual that is in need of therapy or suspected of needing therapy, or an aged or frail individual.
  • a subject can be any human being.
  • any anti-aging treatment is meant to include (but is not limited to) treatments leading to prevention, amelioration or lessening the effects of aging, decreasing or delaying an increase in the biological age, slowing rate of aging; treatment, prevention, amelioration and lessening the effects of frailty or at least one of aging related diseases and conditions or declines or slowing down the progression of such decline (including but not limited to those indicated in Table 1, “Declines”), condition or disease, increasing health span or lifespan, rejuvenation, increasing stress resistance or resilience, increasing rate or other enhancement of recovery after surgery, radiotherapy, disease and/or any other stress, decreasing all-causes or multiple causes of mortality risks or mortality risks related to at least one or at least two of age related diseases or conditions or delaying in increase of such risks, decreasing morbidity risks.
  • the treatment leading to the modulating at least one of biomarkers of aging into more youthful state or slowing down its change into “elder” state is also regarded to be an anti-aging treatment, including but not limited to biomarkers of aging which are visible signs of aging, such as wrinkles, grey hairs etc.
  • an age-related disease or disorder is selected from: atherosclerosis, cardiovascular disease, adult cancer, arthritis, cataracts, osteoporosis, type 2 diabetes, hypertension, neurodegeneration (including but not limited to Alzheimer's disease, Huntington’s disease, and other age-progressive dementias; Parkinson's disease; and amyotrophic lateral sclerosis [ALS]), stroke, atrophic gastritis, osteoarthritis, NASH, camptocormia, chronic obstructive pulmonary disease, coronary artery disease, dopamine dysregulation syndrome, metabolic syndrome, effort incontinence, Hashimoto's thyroiditis, heart failure , late life depression, immunosenescence, myocardial infarction, acute coronary syndrome, sarcopenia, sarcopenic obesity, senile osteoporosis, urinary incontinence etc.
  • ALS amyotrophic lateral sclerosis
  • Aging-related changes in any parameter or physiological metric are also regarded as age-related conditions, including but not limited to aging related change in blood parameters, heart rate, cognitive functions/decline, bone density, basal metabolic rate, systolic blood pressure, heel bone mineral density (BMD), heel quantitative ultrasound index (QUI), heel broadband ultrasound attenuation, heel broadband ultrasound attenuation, forced expiratory volume in 1- second (FEV1), forced vital capacity (FVC), peak expiratory flow (PEF), duration to first press of snap- button in each round, reaction time, mean time to correctly identify matches, hand grip strength (right and/or left), whole body fat-free mass, leg fat-free mass (right and/or left), and time for recovery after any stress (wound, operation, chemotherapy, disease, change in lifestyle etc.).
  • Aging related change in any parameter of organism is also regarded as an aging related condition, including but not limited to aging related change in at least one of the parameter selected from the Table 1 “Declines”.
  • the term “small molecule” means an individual compound with molecular weight less than about 2000 daltons, usually less than about 1500 daltons, more usually less than about 750 daltons, preferably less than about 500 daltons, although molecules larger than 2000 daltons in size will also be included herein.
  • the term “senescence associated disease or disorder” is used interchangeably with “aging related disease or condition”.
  • the term “aging related disease or condition” is used interchangeably with “senescence associated disease or disorder”.
  • COMBINATION WITH OTHER ANTI-AGING DRUGS any compound of this disclosure can be used with other anti-aging drugs or intervetions which will increase anti-aging effects.
  • Some possible geroprotectors include melatonin, carnosine, metformin, nicotinamide mononucleotide (NMN), delta sleep-inducing peptide. NAD+, bASISTM, Rejuvant® LifeTabs®, rapamycin, rapalogs, spermidine and others.
  • NAD+ melatonin
  • NPN nicotinamide mononucleotide
  • delta sleep-inducing peptide NAD+
  • bASISTM Rejuvant® LifeTabs®
  • rapamycin rapalogs
  • spermidine spermidine and others.
  • such pharmaceutical composition is for use as an anti-aging medication or for use for the anti-aging treatment or rejuvenation.
  • compositions for intravenous administration can comprise a sterile isotonic aqueous buffer.
  • the compositions can also include a solubilizing agent.
  • Compositions for intravenous administration can optionally include a local anesthetic such as lignocaine to lessen pain at the site of the injection.
  • a local anesthetic such as lignocaine to lessen pain at the site of the injection.
  • the pharmaceutical composition described herein is administered by infusion, it can be dispensed, for example, with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampule of sterile water for injection or saline can be provided so that the enzyme or enzyme and antioxidant and the carrier can be mixed prior to administration.
  • One of the many possible forms of this invention can be a Lyophilized Concentrate for Intravenous (IV) Injection.
  • the amount of pharmaceutical composition described herein that is effective for treating a corresponding disease or condition can be determined using standard clinical or pharmacokinetic techniques known to those with skill in the art.
  • in vitro or in vivo assays can optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed can also depend on the route of administration, the disease or condition, the seriousness of the corresponding disease or condition being treated, as well as various physical factors related to the individual being treated, and can be decided according to the judgment of a health-care practitioner.
  • any agent or composition of this disclosure in an amount ranging from about 0.05 ⁇ g/kg to about 100 mg/kg of a patient’s body weight, or 0.01 to about 1000 mg/kg of total body weight per day, or from about 0.1 to about 100 mg/kg of total body weight per day, or from about 0.5 to about 15 mg/kg of total body weight per day, or from about 1 mg/kg to about 50 mg/kg of total body weight, which may be administered in one or multiple doses per day or per week or per month or per 6 months or per year or per 3 years or per 8 years or per 12 years or once in a lifetime.
  • this invention is a pharmaceutical composition and formulation, comprising amount of the agent needed for a subject with weight of 50 kg or in some embodiments 75 kg or in some embodiments 90 kg to provide the amount of the agent described above and at least one pharmaceutically acceptable excipient.
  • Equivalent dosages can be administered over various time periods including, but not limited to, about every 2 hours, about every 4 hours, about every 8 hours, about every 12 hours, about every 24 hours, about every 36 hours, about every 48 hours, about every 72 hours, about every week, about every two weeks, about every three weeks, about every month, and about every two months or every 6 months or every year or every 3 years or every 8 years or every 12 years or once in a lifetime or daily lifelong or as decided by practitioner or patient.
  • the number and frequency of dosages corresponding to a completed course of therapy can be determined according to the judgment of a health- care practitioner.
  • the administration in case of toxicity or adverse effects the administration can be suspended or dosage decreased until the toxicity or adverse effects disappear and then the administration and/or dosage can be resumed on the previous level.
  • the pharmaceutical composition and formulations described herein are administered to a subject by any suitable administration route, including but not limited to, parenteral (e.g., intravenous, subcutaneous, intramuscular), intradermal, intraperitoneal, subcutaneous, intranasal, epidural, sublingual, intravaginal, rectal, by inhalation, topical intracerebral, oral, intranasal, buccal, rectal, or transdermal administration routes.
  • parenteral e.g., intravenous, subcutaneous, intramuscular
  • intradermal intraperitoneal
  • subcutaneous intranasal
  • epidural sublingual
  • intravaginal sublingual
  • intravaginal sublingual
  • rectal by inhalation
  • topical intracerebral topical intracerebral
  • oral, intranasal, buccal, rectal or transdermal administration routes.
  • the pharmaceutical composition described herein is administered locally.
  • topical application including but not limited when prodrugs that require esterase activation after uptake may be applicable topically
  • local infusion during surgery by injection, by means of a catheter, by means of a suppository or enema, or by means of an implant, the implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • the pharmaceutical composition described herein is introduced into the central nervous system, circulatory system or gastrointestinal tract by any suitable route, including intraventricular, intrathecal injection, paraspinal injection, epidural injection, enema, and by injection adjacent to a peripheral nerve.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent, or via perfusion in a fluorocarbon or synthetic pulmonary surfactant.
  • the pharmaceutical formulations include, but are not limited to, aqueous fluid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations (e.g., nanoparticle formulations), and mixed immediate and controlled release formulations.
  • the pharmaceutical formulations include a carrier or carrier materials selected on the basis of compatibility with the composition disclosed herein, and the release profile properties of the desired dosage form.
  • exemplary carrier materials include, e.g., binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, and the like.
  • Pharmaceutically compatible carrier materials include, but are not limited to, acacia, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, maltodextrin, glycerine, magnesium silicate, polyvinylpyrrollidone (PVP), cholesterol, cholesterol esters, sodium caseinate, soy lecithin, taurocholic acid, phosphotidylcholine, sodium chloride, tricalcium phosphate, dipotassium phosphate, cellulose and cellulose conjugates, sugars sodium stearoyl lactylate, carrageenan, monoglyceride, diglyceride, pregelatinized starch, and the like.
  • PVP polyvinylpyrrollidone
  • the pharmaceutical formulations further include pH adjusting agents or buffering agents which include acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids, bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane, and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride. Such acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range.
  • the pharmaceutical formulation includes one or more salts in an amount required to bring osmolality of the composition into an acceptable range.
  • Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions, suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
  • the pharmaceutical formulations include, but are not limited to, sugars like trehalose, sucrose, mannitol, maltose, glucose, or salts like potassium phosphate, sodium citrate, ammonium sulfate and/or other agents such as heparin to increase the solubility and in vivo stability of polypeptides.
  • the pharmaceutical formulations include filling agents such as lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.
  • Lubricants and glidants are also optionally included in the pharmaceutical formulations described herein for preventing, reducing or inhibiting adhesion or friction of materials.
  • Exemplary lubricants include, e.g., stearic acid, calcium hydroxide, talc, sodium stearyl fumerate, a hydrocarbon such as mineral oil, or hydrogenated vegetable oil such as hydrogenated soybean oil (Sterotex®), higher fatty acids and their alkali-metal and alkaline earth metal salts, such as aluminum, calcium, magnesium, zinc, stearic acid, sodium stearates, glycerol, talc, waxes, Stearowet®, boric acid, sodium benzoate, sodium acetate, sodium chloride, leucine, a polyethylene glycol (e.g., PEG-4000) or a methoxypolyethylene glycol such as CarbowaxTM, sodium oleate, sodium benzoate, glyceryl behenate, polyethylene glycol, magnesium or sodium lauryl sulfate, colloidal silica such as SyloidTM, Cab-O-Sil®, a starch such as corn starch, silicone
  • Plasticizers include compounds used to soften the microencapsulation material or film coatings to make them less brittle. Suitable plasticizers include, e.g., polyethylene glycols such as PEG 300, PEG 400, PEG 600, PEG 1450, PEG 3350, and PEG 800, stearic acid, propylene glycol, oleic acid, triethyl cellulose and triacetin. Plasticizers can also function as dispersing agents or wetting agents.
  • Solubilizers include compounds such as triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, sodium lauryl sulfate, sodium doccusate, vitamin E TPGS, dimethylacetamide, N-methylpyrrolidone, N- hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropylmethyl cellulose, hydroxypropyl cyclodextrins, ethanol, n-butanol, isopropyl alcohol, cholesterol, bile salts, polyethylene glycol 200-600, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide and the like.
  • Stabilizers include compounds such as any antioxidation agents, buffers, acids, preservatives and the like.
  • Exemplary stabilizers include L-arginine hydrochloride, tromethamine, albumin (human), citric acid, benzyl alcohol, phenol, disodium biphosphate dehydrate, propylene glycol, metacresol or m-cresol, zinc acetate, polysorbate-20 or Tween® 20, or trometamol.
  • Suspending agents include compounds such as polyvinylpyrrolidone, e.g., polyvinylpyrrolidone K12, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30, vinyl pyrrolidone/vinyl acetate copolymer (S630), polyethylene glycol, e.g., the polyethylene glycol can have a molecular weight of about 300 to about 6000, or about 3350 to about 4000, or about 7000 to about 5400, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, hydroxymethylcellulose acetate stearate, polysorbate-80, hydroxyethylcellulose, sodium alginate, gums, such as, e.g., gum tragacanth and gum acacia, guar gum, xanthans, including xanthan gum, sugars, cellulosics, such as, e
  • Surfactants include compounds such as sodium lauryl sulfate, sodium docusate, Tween 60 or 80, triacetin, vitamin E TPGS, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic® (BASF), and the like.
  • compounds such as sodium lauryl sulfate, sodium docusate, Tween 60 or 80, triacetin, vitamin E TPGS, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic® (BASF), and the like.
  • Pluronic® Pluronic®
  • Additional surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil, and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40. Sometimes, surfactants is included to enhance physical stability or for other purposes.
  • Viscosity enhancing agents include, e.g., methyl cellulose, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, hydroxypropylmethyl cellulose acetate stearate, hydroxypropylmethyl cellulose phthalate, carbomer, polyvinyl alcohol, alginates, acacia, chitosans and combinations thereof.
  • Wetting agents include compounds such as oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, sodium docusate, sodium oleate, sodium lauryl sulfate, sodium doccusate, triacetin, Tween 80, vitamin E TPGS, ammonium salts and the like.
  • Methods and Uses [14] Described herein are methods of treating or preventing an age-related disease or disorder or other anti-aging treatment comprising administering to a subject in need thereof an agent or pharmaceutical composition described herein.
  • the compound selected from the table 1 ab and Table 1c and combinations of this disclosure are useful for changing selected biomarkers related to aging or mortality risks into a younger state and thus reducing the risks of mortality, including but not limited to biomarkers described in Table 2, https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5611985/ or on the website http://mortalitypredictors.org and the publications cited there on blood predictors of mortality or in any other source. [17] Every web link cited in this application, in case of inaccessibility can be retrieved via https://web.archive.org or similar internet archive services.
  • this invention is a kit, comprising a compound selected from the table 1 ab and Table 1c or other agent, disclosed or described in this disclosure, or composition disclosed in this application or its functional or structural analog or prodrug and the notice, description or instruction for its use for anti-aging treatment, optionally comprising at least the medication labeling information.
  • this invention is a kit, comprising a compound selected from the table 1 ab and Table 1c or other agent, disclosed or described in this disclosure, or composition disclosed in this application or its functional or structural analog or prodrug and the notice, description or instruction for its use by human or by other animal subject in a dosage and regimen to maintain concentration of such compound in blood of such subject in 1 ⁇ M level at 30% of time, or from 0,5 to 5 ⁇ M level at from around 1% of time to around 100% of time, or from 0,5 to 5 ⁇ M level at from around 10% of time to around 50% of time, or from 0,05 to 5 ⁇ M level or at least one other level described in this disclosure.
  • the kit of this invention can be made of any material such as paper, plastic, steel, glass, etc, including but not limited to materials used in the art for kits for medications.
  • this invention is an compound (agent) of this disclosure with the mentioned notice, description or instruction attached to such agent or imprinted or drawn or in any other way displayed on such agent or in any other way associated with such agent (e.g. in machine readable form).
  • One of the primary purposes of some aspects of his invention is to provide medication for anti-aging treatment.
  • the contents and appearance of such notice, description or instruction is regulated by the respective national or international rules regarding labeling of medication, incorporated here by reference or such notice, description or instruction comprise at least part or optionally most of the or optionally all the information required by applicable labeling regulations.
  • the notice, description or instruction including but not limited to labeling means (e.g., treatment and/or operation guidelines) can be provided in any form that conveys the requisite information.
  • Instruction means can be audio, for example spoken word, recorded in analog or digital form (e.g., audio recording), or received and/or transmitted in analog or digital form (e.g., by telephone, conference call, or audio signal transmitted over a network).
  • Such information can also be visual or video, for example hard-copy (e.g., as a manual, recorded medium, booklet, leaflet, book and the like) or soft-copy (e.g., recorded in analog or digital form as a file recorded on an magnit, electronic, optical, or computer readable medium such as a DVD, disk drive, CD-ROM and the like).
  • instruction means can be interactive or real-time (e.g., a teleconference or internet chat or chat bot).
  • Some kits or agents of this invention can include printed or made in any other way instructions to inform the user of the steps required to properly use it.
  • agents and kits, of this invention include a label configured to be coupled to respective agent and kits of this invention.
  • the label includes a first surface and a second surface.
  • the first surface can be coupled to an outer surface of agents and kits of this invention.
  • the first surface can include an adhesive.
  • the second surface can include include a textual indicia, such as, for example, a description of the of agents and kits of this invention , a mark indicating its manufacturer or distributor and/or an instruction associated with the use of such of agents and kits of this invention.
  • the label can further include an electronic circuit system configured to output an electronic signal.
  • the electronic signal can include an instruction associated with the use of the mediums, kits, devices or agents of this invention. [26]
  • the instruction is an instruction for use as anti-aging medication.
  • the instruction is an instruction for use as medication for a treating or preventing an age- related disease or disorder.
  • the notice, description or instruction including but not limited to labeling can be shown on the lenses, computer glasses, transmitted via brain computer interface or by any other means or can be encoded by the Quick Response Code or any other machine readable form.
  • the notice, description or instruction, including but not limited to labeling can be implemented in digital electronic circuitry, or in computer firmware, hardware, software, or in combinations thereof.
  • the implementation can be as a computer program product, e.g., a computer program tangibly embodied in an information carrier, e.g., in a machine-readable storage device or in a propagated signal, for execution by, or to control the operation of, data processing apparatus, e.g., a programmable processor, a computer, or multiple computers.
  • a computer program can be recorded in any form of programming language, including compiled or interpreted languages, and the computer program can be deployed in any form, including as a stand-alone program or as a subroutine, element, or other unit suitable for use in a computing environment.
  • a computer program can be deployed to be executed on one computer or on multiple computers at one site or several sites.
  • the notice, description or instruction, including but not limited to labeling can be performed by one or more programmable processors executing a computer program to perform functions of the invention by operating on input data and generating output. It can also be performed by, and an apparatus can be implemented as, special purpose logic circuitry, e.g., an FPGA (field programmable gate array) or an ASIC (application-specific integrated circuit). Subroutines can refer to portions of the computer program and/or the processor/special circuitry that implements that functionality.
  • the dosage levels and mode of administration will be dependent on a variety of factors such as the treatment used, the device, the active agent, the context of use (e.g., the patient to be treated), and the like.
  • the compound s of this invention are known in the art as well as the process of its manufacturing.
  • the compound s of this invention can also be acquired from commercial sources.
  • the one or two or more biomarkers (optionally - with associated measurement units in plasma) which will be changed into more youthful state as a result of administration of compound or combination of this invention is a biological age characteristic.
  • biological age characterizes the health status of the subject.
  • a compound selected from the table 1 ab and Table 1c or ther agents and combinations of this disclosure are administered by aged subject.
  • the biological age determination approach is described in prior art, including but not limited to any of the following publications https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5514388/ . https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4931851/, https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5514388/ and corresponding references related to blood based biological age determination.
  • the biological age is understood as the distance measured along a continuous trajectory consisting of distinct phases, each corresponding to subsequent human life stages as described in more details in https://www.biorxiv.org/content/biorxiv/early/2017/03/09/186569.full.pdf [37]
  • this at least one of the compound , composition or combination of this disclosure reduces mortality risk or high mortality risk in about 1 month, in about 3 months, in about 6 months, in about 1 year, from about 1 month to about 6 months, from about 1 month to about 1 year, from about 1 year to about 3 years, from about 3 years to about 5 years, from about 5 years to about 8 years, from about 5 years to about 10 years, in about 5 years, in about 10 years, in about 15 years.
  • mortality risk is a risk of dying from age related condition or disease. In some embodiments, mortality risk is all cause mortality risk.
  • biomarkers of mortality and its critical volumes are described in prior art, including but not limited to https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4899173/ , https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4454670/ , https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5334528/ Maximus Peto, Carlos De la Guardia, Ksenia Winslow, Andrew Ho, Kristen Fortney, & Eric Morgen.
  • a compound selected from the table 1 ab and Table 1c composition or combination of this disclosure reduces morbidity risk in about 1 month, in about 3 months, in about 6 months, in about 1 year, from about 1 month to about 6 months, from about 1 month to about 1 year, from about 1 year to about 3 years, from about 3 years to about 5 years, from about 5 years to about 8 years, from about 5 years to about 10 years, in about 5 years, in about 10 years, in about 15 years.
  • morbidity risk is a risk of acquiring an age related condition or disease.
  • this invention is a method, comprising administration of the compound selected from the table 1 ab and Table 1c , composition or combination of this disclosure, for the at least one of the selected from the group consisting of: reducing biological age of the patient, improving of at least one aging biomarker, alleviating of at least one age related deficit or disease, improving of at least one of rejuvenation marker, treating frailty, increasing health span or life span, providing an anti-aging treatment.
  • this invention is at least one of the of the compounds selected from the table 1 ab and Table 1c , composition or combination of this disclosure useful for treatment and prevention of disease selected from the group: type 2 diabetes, age-related cardiovascular diseases, including but not limited to ischemic heart disease and stroke, metabolic syndrome, COPD, Alzheimer’s disease etc., including but not limited those mentioned in this disclosure or at least one of the aging related declines.
  • treatment by the compound selected from the table 1 ab and Table 1c may include treatment of any disease or condition which is fully or partially mediated by the induction or maintenance of a non-proliferating or senescent state in a cell or a population of cells in a subject.
  • Non-limiting examples include age-related tissue or organ decline which may lack visible indication of pathology, or overt pathology such as a degenerative disease or a function-decreasing disorder.
  • a degenerative disease or a function-decreasing disorder For example, Alzheimer's disease, Parkinson's disease, cataracts, macular degeneration, glaucoma, atherosclerosis, acute coronary syndrome, myocardial infarction, stroke, hypertension, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), osteoarthritis, type 2 diabetes, obesity, fat dysfunction, coronary artery disease, cerebrovascular disease, periodontal disease, and cancer treatment-related disability such as atrophy and fibrosis in various tissues, brain and heart injury, and therapy-related myelodysplastic syndromes.
  • IPF idiopathic pulmonary fibrosis
  • COPD chronic obstructive pulmonary disease
  • osteoarthritis type 2 diabetes, obesity, fat dysfunction, coronary
  • an age- related pathology may include an accelerated aging disease such as Hutchinson-Gilford progeria syndrome, Werner syndrome, Cockayne syndrome, exroderma pigmentosum, ataxia telangiectasia, Fanconi anemia, dyskeratosis congenital, aplastic anemia, idiopathic pulmonary fibrosis, and others.
  • a method of identifying an age-related disease or condition as described herein may include detecting the presence of senescent cells.
  • a senescence-associated pathology that can be treated by compound selected from the table 1 ab and Table 1c may include any disease or condition which is fully or partially mediated by the induction or maintenance of a non- proliferating or senescent state in a cell or a population of cells in a subject.
  • Non-limiting examples include cardiovascular diseases such as angina, aortic aneurysm, arrhythmia, brain aneurysm, cardiac diastolic dysfunction, cardiac fibrosis, cardiac stress resistance, cardiomyopathy, carotid artery disease, coronary thrombosis, endocarditis, hypercholesterolemia, hyperlipidemia, mitral valve prolapsed, and peripheral vascular disease; inflammatory or autoimmune diseases such as herniated intervertebral disc, inflammatory bowel disease, kyphosis, oral mucositis, lupus, interstital cystitis, scleroderma, and alopecia; neurodegenerative diseases such as dementia,Huntington's disease, motor neuron dysfunction, age-related memory decline, and depression/mood disorders; metabolic diseases such as diabetic ulcer and metabolic syndrome; pulmonary diseases such as age-related loss of pulmonary function, asthma, bronchiectasis, cystic fibrosis, emphysema, and age
  • a method for treating a senescence associated disease or disorder in a subject comprising administering to the subject a compound selected from the table 1 ab and Table 1c or combination, combination, comprising such compound, ; wherein the senescence associated disease or disorder is not a cancer, wherein such compound or combination is administered during a treatment course of 1-7 days every 0.5-12 months; provided that if the senescence associated disease or disorder is a senescence associated metabolic disorder, such compound or combination is administered during a treatment course of 1-7 days every 4-12 months.
  • such compound or combination is administered once every 0.5-12 months; provided that if the senescence associated disease or disorder is a senescence associated metabolic disorder, such compound or combination is administered once every 4- 12 months.
  • the aging related disease or senescent cell-associated disease or disorder is a cardiovascular disease or disorder, inflammatory disease or disorder, a pulmonary disease or disorder, a neurological disease or disorder.
  • the cardiovascular disease or disorder is atherosclerosis.
  • the inflammatory disease or disorder is osteoarthritis.
  • the pulmonary disease or disorder is idiopathic pulmonary fibrosis or chronic obstructive pulmonary disease.
  • the neurological disease or disorder is selected from mild cognitive impairment; motor neuron dysfunction; Alzheimer's disease; Parkinson's disease; and macular degeneration.
  • the senescence associated metabolic disease or disorder is selected from diabetes, metabolic syndrome, and obesity.
  • the aging related disease or senescence-associated disease or disorder is a dermatological disease or disorder is selected from eczema, psoriasis, hyperpigmentation, nevi, rashes, atopic dermatitis, urticaria, diseases and disorders related to photosensitivity or photoaging, rhytides; pruritis; dysesthesia; eczematous eruptions; eosinophilic dermatosis; reactive neutrophilic dermatosis; pemphigus; pemphigoid; immunobullous dermatosis;fibrohistocytic proliferations of skin; cutaneous lymphomas; and cutaneous lupus.
  • a method for treating a senescence- associated metabolic disease or disorder in a subject comprising administering to the subject a compound selected from the table 1 ab and Table 1c or combination, comprising such compound , wherein such compound or combination is administered during a treatment course of 1-7 days every 4-12 months, and wherein the metabolic disease or disorder is selected from diabetes, metabolic syndrome, and obesity.
  • such compound or combination is administered once every 4-12 months.
  • the senescent cell is selected from a senescent fibroblast, a senescent pre- adipocyte, a senescent epithelial cell, a senescent chondrocyte, a senescent neuron, and a senescent endothelial cell.
  • the senescent cell is a senescent pre-adipocyte.
  • a method for treating, reducing the likelihood of occurrence of, or delaying onset of age related disease or decline or a senescent cell-associated disease or disorder in a subject who has an age related disease or decline a senescent cell-associated disease or disorder or who has at least one predisposing factor for developing the senescent cell-associated disease or disorder comprising administering to the subject a compound selected from the table 1 ab and Table 1c or combination, comprising such compound, , thereby promoting death of the senescent cell, wherein the senescent cell-associated disease or disorder is a cardiovascular disease or disorder, inflammatory disease or disorder, a pulmonary disease or disorder, a neurological disease or disorder, with the proviso that if the subject has a cancer, and such compound is not a primary therapy for treating the cancer, and wherein the such compound or composition is administered once every 0.5-12 months.
  • a method for treating, reducing the likelihood of occurrence of, or delaying onset of a senescent cell-associated disease or disorder in a subject who has a senescent cell- associated disease or disorder or who has at least one predisposing factor for developing the senescent cell- associated disease or disorder, comprising administering to the subject a senolytic combination comprising (a) a first agent that alters either one or both of a cell survival signaling pathway and an
  • a method is provided for killing a senescent cell comprising contacting the senescent cell and a compound selected from the table 1 ab and Table 1c or combination, comprising such compound, thereby promoting death of the senescent cell, wherein the senescence cell is present in a subject who has a senescent cell-associated disease or disorder or who has at least one predisposing factor for developing the senescent cell-associated disease or disorder, with the proviso that if the subject has
  • the senescent cell-associated disease or disorder is selected from atherosclerosis; osteoarthritis; idiopathic pulmonary fibrosis; chronic obstructive pulmonary disease; mild cognitive impairment; motor neuron dysfunction; Alzheimer's disease; Parkinson's disease; and macular degeneration.
  • a method for killing a senescent cell comprising contacting the senescent cell and compound selected from the table 1 or combination, comprising such compound, , thereby promoting death of the senescent cell, wherein the senescence cell is present in a subject who has a senescent cell-associated disease or disorder or who has at least one predisposing factor for developing the senescent cell-associated disease or disorder, wherein the senescent cell-associated disease or disorder is a metabolic disorder selected from diabetes, metabolic syndrome, and obesity, and wherein the combination is administered once every 4-12 months.
  • a method for treating or reducing the likelihood of occurrence of atherosclerosis in a subject who has atherosclerosis or who has at least one predisposing factor for developing atherosclerosis comprising administering to the subject a compound selected from the table 1 ab and Table 1c or combination, comprising such compound, thereby promoting death of the senescent cell, wherein the combination is administered once every 0.5-12 months, and wherein the first and second agents are different.
  • the senescent cell is selected from a senescent fibroblast, a senescent pre-adipocyte, a senescent epithelial cell, a senescent chondrocyte, a senescent neuron, a senescent smooth muscle cell, a senescent mesenchymal cell, a senescent macrophage, and a senescent endothelial cell.
  • the senescent cell is a senescent pre- adipocyte.
  • this invention is a method, including but not limited to method of testing of efficacy of compound, composition or combination of this disclosure, comprising the checking in the subject treated by such therapy at least one of the following: checking biological age of the patient, at least one aging biomarker, at least one age related deficit or disease, at least one of rejuvenation marker, frailty, health span or life span, or any other marker or parameter reasonable for checking in testing of anti-aging therapy efficacy.
  • this invention is a method of anti-aging treatment, comprising administering by subject at least one compound, composition or combination of this disclosure, and repeating administration in case biological age increased for more than 1 year, more than 3 years, more than 8 years, more than 10 years.
  • the dosage and regimen for implementing disclosed method is defined to keep biological age at the reasonably minimum level, or close to the age that subject had at the time of first treatment.
  • checking of efficacy of compound, composition or combination of this disclosure and measurement of markers or symptoms of related diseases or conditions is conducted in 1 month after the infusion of treated plasma or administration of therapy in therapeutically effective amount, in 3 months, in 6 months, in 12 months, in 18 months, in 24 months or in 36 months after such infusion, or in around such date, or in date reasonably defined by the practitioner based on the parameter being measured and other factors known to the expert in the field .
  • any of the compound s and combinations of this disclosure can be provided in formulations defined by the expert skilled in the art, including but not limited those known in the art and those suggested in the examples.
  • compounds selected from the table 1 ab and Table 1c or their structural or functional analogs or prodrugs and other agents of this disclosure to be useful for anti-aging or treatment of age related disease or other disease indicated in this disclosure shall be administered in the same dosages as such agents are effective in their primary indication (Primary indication effective dosage or PIED), which is known in the art.
  • Primary indication effective dosage or PIED Primary indication effective dosage
  • the dosage for the agent of this disclosure should be at least 100 times less or least 50 times less or at least 10 times less or at least 5 times less, or at least 2 times less, or at least 50% less, or at least 25% less, or at least 10% less, or at least 5 times more, or at least 2 times more, or at least 50% more, or at least 25% more, or at least 10% more, or at least 10 times more, or at least 100 times more than PIED to have anti-aging therapeutic effect.
  • to have anti-aging therapeutic effect such agent shall be administered at maximum tolerated dose.
  • PIED is a dosage which was tested in Phase 1. In some embodiments, PIED is a dosage which proved to be safe in Phase 1. [47] In some embodiments, the dosage for the compound selected from the table 1 ab and Table 1c should be selected from the group consisting of: about at least 1000 times less at least 100 times less or least 50 times less or at least 10 times less or at least 5 times less, or at least 2 times less, or at least 50% less, or at least 25% less, or at least 10% less, than maximum tolerated dose to have anti-aging therapeutic effect or to be effective against aging, frailty, aging related disease or condition or other disease or condition mentioned in this application.
  • this invention is a tangible medium comprising a computer program, which, when executed, causes a medium to perform a method comprising: attribution to the information about a subject an information about an anti-aging treatment related to compound, composition or combination of this disclosure, optionally further comprising attributing to the information about patient before or after or before and after the treatment to information about checking of at least one selected from the group: biological age of the patient, at least one aging biomarker, at least one age related deficit or disease or its symptom, at least one of rejuvenation marker, frailty, health span or life span.
  • a prophetic example of such tangible medium could be a APPLE TM 2014 MACBOOK AIRTM 13" intelTM i5 with Microsoft TM Excel TM installed and executed on it, wherein to patient with name John Junior Smith (born 2 Jan 1937) the information about using compound selected from the table 1 ab and Table 1c or their pharmaceutically acceptable salt or any combination of it or compounds or , optionally, having the similar SAR characteristics in therapeutically effective amount is attributed in the sense that is logically linked as an information in Excel table (in this example attribution is realized as placing the information about treatment by such drug in the same line in the file with the name and ID of the patient to whom such treatment is prescribed) and allows easy finding of patients to whom such treatment is prescribed and other processing of such information.
  • processors suitable for the execution of a computer program related to this invention include, by way of example, both general and special purpose microprocessors, and any one or more processors of any kind of digital computer.
  • a processor receives instructions and data from a read-only memory or a random access memory or both.
  • the essential elements of a computer are a processor for executing instructions and one or more memory devices for storing instructions and data.
  • a computer also includes, or be operatively coupled to receive data from or transfer data to, or both, one or more mass storage devices for storing data, e.g., magnetic, magneto-optical disks, or optical disks. Data transmission and instructions can also occur over a communications network.
  • Information carriers suitable for embodying computer program instructions and data include all forms of non-volatile memory, including by way of example semiconductor memory devices, e.g., EPROM, EEPROM, and flash memory devices; magnetic disks, e.g., internal hard disks or removable disks; magneto-optical disks; and CD-ROM and DVD-ROM disks.
  • semiconductor memory devices e.g., EPROM, EEPROM, and flash memory devices
  • magnetic disks e.g., internal hard disks or removable disks
  • magneto-optical disks e.g., CD-ROM and DVD-ROM disks.
  • the processor and the memory can be supplemented by, or incorporated in special purpose logic circuitry.
  • this invention is a tangible medium comprising a computer program, which, when executed, causes a device to perform a method comprising: attribution to the information regarding compound, composition or combination of this disclosure, an information about anti-aging treatment.
  • this invention is a method, method comprising: attribution to the information about a subject an information about an anti-aging treatment related to compound selected from the table 1 ab and Table 1c , composition or combination of this disclosure, optionally further comprising attributing to the information about patient before or after or before and after the treatment to information about checking of at least one selected from the group: biological age of the patient, at least one aging biomarker, at least one age related deficit or disease, at least one of rejuvenation marker, frailty, health span or life span.
  • such method is a computer implemented method.
  • this invention is a method, the method of this invention, comprising attribution of information executed on the medium of this invention and described in corresponding part of this disclosure related to such medium.
  • this invention is a tangible medium or computer system or processor, comprising a computer program, which, when executed, causes a medium to perform a method comprising attribution of information described in this disclosure.
  • this invention is an apparatus to execute method described in this disclosure, the apparatus comprising the processor comprising the tangible medium described in this disclosure
  • dose for anti-aging effect of the compound selected from the table 1 ab and Table 1c is about 500 mg (one tablet) 3-4 times a day.
  • dose of the compound selected from the table 1 ab and Table 1c is from about 50 mg to about 4000 mg. In some embodiment, dose of the compound selected from the table 1 ab and Table 1c is from about 10 mg to about 4000 mg. In some embodiment, dose of the compound selected from the table 1 ab and Table 1c is from 100 mg to 2000 mg. In some embodiment, dose of the compound selected from the table 1 ab and Table 1c is from about 1 mg to about 100 mg. In some embodiment, dose of the compound selected from the table 1 ab and Table 1c is from about 10 mg to about 50 mg. In some embodiments, this invention is a pharmaceutical composition or formulation comprising single dosage in the amount described above and at least one pharmaceutically acceptable excipient.
  • the compound selected from the table 1 ab and Table 1c is administered intramuscularly.
  • the single dose for all age groups is 8-16 mg/kg body weight according to the following scheme: 16-31 kg body weight - 250 mg the compound selected from the table 1 ab and Table 1c (1/2 ml); 32-46 kg body weight - 500 mg the compound selected from the table 1 ab and Table 1c (1 ml); 47-62 kg body weight – from about 500 to about 750 mg the compound selected from the table 1 ab and Table 1c (1-1.5 ml); and over 63 kg body weight - 750-1000 mg the compound selected from the table 1 ab and Table 1c (1.5-2 ml).
  • the dose can be repeated in 6-8 hours. In some embodiments, the dose in adults is 250-500 mg (1/2 -1 tablet) 2 or 3 times daily. In some embodiments, maximal 24-hour dose is about amount selected from the group consisting of: from about 0,01 mg to about 0,1 mg, from about 0,1 mg to about 1 mg, from about 1 mg to about 10 mg, from about 10 mg to about 100, from about 100 mg to about 1000 mg, from about 1 g to about 10 g, from about 0,01 mg to about 10g. In some embodiments the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c is administered from 5 mg/m2 to 200 mg/m2.
  • the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c is administered at a dose shown above or of therapeutic efficacy, 2-3 times daily. [59] In some embodiments, the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c can be administered as a shot into the fatty part of the skin. In some embodiments, The compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c can be administered as an infusion into a vein over a period of time.
  • the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c can be administered in the dosage from about 0,1 mg/m2/day to about 1000 mg/m2/day or from about 1 mg/m2/day to about 100 mg/m2/day via subcutaneous injection or IV infusion for about from 1 day to about 365 days.
  • the administration of the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c should be repeated cycles about every 4 weeks, or about every 6 weeks, about every 8 weeks or about every 16 weeks, about every 1 year, about every 2 years, about every 5 years, about every 8 years.
  • the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or its pharmaceutically acceptable salt or any combination of it or compounds or , optionally, having the similar SAR characteristics or any of its combination to achieve anti-aging effect can be administered by human or by other animal subject in a dosage and regimen to create or in some other embodiment to maintain a concentration of such compound in blood of such subject in 0.01 ⁇ M level, or in 0.03 ⁇ M level, or in 0.1 ⁇ M level, or in 0.02 ⁇ M level, or in 0.04 ⁇ M level, or in 0.08 ⁇ M level, or in 0.1 ⁇ M level, or in 0.2 ⁇ M level, or in 0.4 ⁇ M level, or in 0.8 ⁇ M, level, or in 1 ⁇ M level, or in 2 ⁇ M level, or in 4 ⁇ M level, or in 8 ⁇ M level, or in 10 ⁇ M level, or in 12 ⁇ M level, or in 14 ⁇ M level, or in 18 ⁇ M level, or
  • this invention is a delivery device or dosing device delivering the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or their pharmaceutically acceptable salt or any combination of it or compounds or , optionally, having the similar SAR characteristics or any of its combination, and enabling maintaining a concentration of such compound in blood of such subject in 1 ⁇ M level at 30% of time, or from 0,5 to 5 ⁇ M level at from around 1% of time to around 100% of time, or from 0,5 to 5 ⁇ M level at from around 10% of time to around 50% of time or concentration described in this disclosure for a time described in this disclosure.
  • this invention is implantable medical device for controlled delivery of therapeutic agents of this disclosure.
  • One of many examples of this invention can have a titanium reservoir, and a porous titanium oxide based membrane to control the rate of release of the therapeutic agent of this invention.
  • the reservoir can contain a formulation of the active agent, including a stabilizer for the active agent, wherein the stabilizer is provided in an extended release configuration.
  • Further details on some of the examples of devices for delivery of compounds of this invention to maintain the a concentration of such compound in blood of such subject in 1 ⁇ M level at 30% of time, or from about 0,01 to about 500 ⁇ M level at from around 1% of time to around 100% of time, or from 0,5 to 5 ⁇ M level at from around 10% of time to around 50% of time or a concentration described in this disclosure for a time described in this disclosure can be found e.g.
  • this invention is a formulation comprising the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or their pharmaceutically acceptable salt or any combination of it or compounds or , optionally, having the similar SAR characteristics or any of its combination, enabling maintaining a concentration of such compound in blood of subject in 0,5 ⁇ M 1 ⁇ M level at 30% of time, or from about 0,05 ⁇ M to about 500 ⁇ M level at from around 1% of time to around 100% of time, or from 0,5 ⁇ M to 5 ⁇ M level at from around 10% of time to around 50% of time or a concentration described in this disclosure for a time described in this disclosure.
  • this invention is a slow release formulation or prodrug, comprising the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or their pharmaceutically acceptable salt or any combination of it or compounds or , optionally, having the similar SAR characteristics or any of its combination, enabling maintaining a concentration of such compound in blood of subject in 1 ⁇ M level at 30% of time, or from 0,5 to 5 ⁇ M level at from around 1% of time to around 100% of time, or from about 0,05 to about 500 ⁇ M level at from around 10% of time to around 50% of time or a concentration described in this disclosure for a time described in this disclosure.
  • such slow release formulation or prodrug is for anti-aging use.
  • this invention is a pharmaceutical composition for oral administration, including but not limited to tablet, capsule, suspension, drink etc., comprising the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c – about 100 mg, or about 50 mg, or about 75 mg, or from about 10 mg to about 150 mg, or from about 20 mg to about 100 mg, or from about 1 mg to about 150 mg, or from about 0.01 mg to about 1000 mg.
  • this invention is a pharmaceutical composition, comprising the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c – about 100 mg, or about 50 mg, or about 75 mg, or from about 10 mg to about 150 mg, or from about 20 mg to 100 mg, or from 1 mg to 150 mg, in some embodiments, this invention such pharmaceutical composition is for anti-aging use.
  • the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c should be administered once daily until disease progression stopped or at least one symptom of age related condition is alleviated or no longer tolerated by the patient.
  • this invention is a pharmaceutical composition for oral administration, including but not limited to tablet, capsule, suspension, drink etc., comprising the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or their pharmaceutically acceptable salt or any combination of it or compounds or , optionally, having the similar SAR characteristics, in some embodiments, this invention such pharmaceutical composition is for anti-aging use.
  • this invention is a pharmaceutical composition and formulation, comprising amount of the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c or their pharmaceutically acceptable salt or any combination of it or compounds or , optionally, having the similar SAR characteristics in the amount needed for a single dose to provide a concentration described in this disclosure for a time described in this disclosure calculated for a subject with weight of 50 kg or in some embodiments 75 kg or in some embodiments 90 kg and at least one pharmaceutically acceptable excipient.
  • Any of the compound selected from the table 1 and combinations of this disclosure can be provided in formulations defined by the expert skilled in the art, including but not limited those known in the art and those suggested in the examples.
  • this invention the new use of the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c wherein a new use is a treatment of viral disease, decreasing a mortality of elderly and/ or frail patients, reduction of a mortality of elderly and/ or frail patients from viral disease, decreasing a mortality of elderly and/ or frail patients from covid-19, decreasing a mortality of elderly and/ or frail patients from influenza, decreasing a mortality of elderly and/ or frail patients from disease, caused by SARS-CoV, SARS-CoV-2 (Severe acute respiratory syndrome coronavirus 2) or other coronavirus, reduction of pulmonary fibrosis caused by viral infection of SARS-CoV, reduction of pulmonary fibrosis caused by viral infection of SARS-CoV-2, reduction of virus titer of SARS-CoV, reduction of virus titer of SARS-CoV-2 [73] In some embodiments, this invention the new use of the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c wherein
  • the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c is useful for treatment and prevention of disease or condition selected from the group: accelerated aging associated with cancer treatment, cancer survival, cognitive impairment associated with cancer treatment and survival, trauma, stroke, HIV, Down syndrome, schizophrenia, condition associated with the cancer treatment, other diseases and conditions associated with increased frailty.
  • the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c is useful as vaccine adjuvant.
  • vaccines examples are vaccine against influenza A, SARS, SARS cov-2, Tetanus and Diphtheria (Td) vaccine; Tetanus, Diphtheria, and Pertussis (Tdap) vaccine; Shingles vaccine; Pneumococcal polysaccharide vaccine; Influenza (flu) vaccine. and vaccines from other infections.
  • such vaccine adjuvant improves immune response to vaccine.
  • the compound selected from the group consisting of all compounds listed in table 1 ab and Table 1c improves immune response to vaccine declined because of aging or frailty or stress or some other disease.
  • each treatment course is no longer than (a) one month, or (b) no longer than two months, or (c) no longer than 3 months. In a specific embodiment, each treatment course is no longer than (a) 5 days, (b) 7 days, (c) 10 days, (d) 14 days, or (e) 21 days.
  • the compound selected from the table 1 ab and Table 1c is administered every 2 nd day or every 3 rd day of each treatment course. In a specific embodiment, the treatment course is one day, two days, three days, or four days. In another specific embodiment, the compound selected from the table 1 ab and Table 1c is administered daily during each treatment course.
  • the non-treatment interval is at least two weeks, at least one month, at least 2 months, at least 3 months, at least 6 months, at least 9 months, or at least 1 year.
  • the treatment course is one
  • the senescence-associated disease or disorder is a cardiovascular disease selected from atherosclerosis, angina, arrhythmia, cardiomyopathy, congestive heart failure, coronary artery disease, carotid artery disease, endocarditis, coronary thrombosis, myocardial infarction, hypertension, aortic aneurysm, cardiac diastolic dysfunction, hypercholesterolemia, hyperlipidemia, mitral valve prolapsed, peripheral vascular disease, cardiac stress resistance, cardiac fibrosis, brain aneurysm, and stroke.
  • the senescence-associated disease or disorder is an inflammatory or autoimmune disease or disorder selected from osteoarthritis, osteoporosis, oral mucositis, inflammatory bowel disease, kyphosis, and herniated intervertebral disc.
  • the senescence-associated disease or disorder is a neurodegenerative disease selected from Alzheimer's disease, Parkinson's disease, Huntington's disease, dementia, mild cognitive impairment, and motor neuron dysfunction.
  • the senescence-associated disease or disorder is a metabolic disease selected from diabetes, diabetic ulcer, metabolic syndrome, and obesity.
  • the senescence-associated disease or disorder is a pulmonary disease selected from pulmonary fibrosis, chronic obstructive pulmonary disease, asthma, cystic fibrosis, emphysema, bronchiectasis, and age-related loss of pulmonary function.
  • the senescence-associated disease or disorder is an eye disease or disorder selected from macular degeneration, glaucoma, cataracts, presbyopia, and vision loss.
  • the senescence- associated disease or disorder is an age-related disorder selected from renal disease, renal failure, frailty, hearing loss, muscle fatigue, skin conditions, skin wound healing, liver fibrosis, pancreatic fibrosis, oral submucosa fibrosis, and sarcopenia.
  • the senescence-associated disease or disorder is a dermatological disease or disorder is selected from eczema, psoriasis, hyperpigmentation, nevi, rashes, atopic dermatitis, urticaria, diseases and disorders related to photosensitivity or photoaging, rhytides; pruritis; dysesthesia; eczematous eruptions; eosinophilic dermatosis; reactive neutrophilic dermatosis; pemphigus; pemphigoid; immunobullous dermatosis; fibrohistocytic proliferations of skin; cutaneous lymphomas; and cutaneous lupus.
  • the senescence-associated disease or disorder is atherosclerosis; osteoarthritis; pulmonary fibrosis; hypertension, or chronic obstructive pulmonary disease.
  • the compound selected from the table 1 ab and Table 1c is administered directly to an organ or tissue that comprises the senolytic cells.
  • the compound selected from the table 1 ab and Table 1c is combined with at least one pharmaceutically acceptable excipient to formulate a pharmaceutically acceptable composition to provide timed-release of the Compound selected from the table 1 ab and Table 1c .
  • the compound selected from the table 1 ab and Table 1c is administered as a bolus infusion.
  • the senescence-associated disease or disorder is osteoarthritis and the compound selected from the table 1 ab and Table 1c is administered directly into the osteoarthritic joint. In another specific embodiment, the compound selected from the table 1 ab and Table 1c is administered intra-articularly to the osteoarthritic joint. In another specific embodiment, the compound selected from the table 1 ab and Table 1c is administered topically, transdermally, or intradermally. In another specific embodiment, the senescence-associated disease or disorder is osteoarthritis and the compound selected from the table 1 ab and Table 1c induces production of Type II collagen in a joint.
  • the senescence-associated disease or disorder is osteoarthritis and the compound selected from the table 1 ab and Table 1c inhibits erosion of a proteoglycan layer in a joint.
  • the senescence-associated disease or disorder is osteoarthritis and the compound selected from the table 1 ab and Table 1c inhibits erosion of a bone of a joint.
  • pulmonary fibrosis is idiopathic pulmonary fibrosis.
  • the compound selected from the table 1 ab and Table 1c reduces the amount of fibrotic pulmonary tissue in the lung.
  • the compound selected from the table 1 ab and Table 1c is administered intranasally, by inhalation, intratracheally, or by intubation.
  • the senescence associated disease or disorder is atherosclerosis, and wherein the compound selected from the table 1 ab and Table 1c increases the stability of atherosclerotic plaque.
  • the senescence associated disease or disorder is atherosclerosis, and wherein the compound selected from the table 1 ab and Table 1c inhibits formation of atherosclerotic plaque in a blood vessel of the subject.
  • the senescence associated disease or disorder is atherosclerosis, and wherein the compound selected from the table 1 ab and Table 1c reduces the lipid content of an atherosclerotic plaque in a blood vessel of the subject.
  • the senescence associated disease or disorder is atherosclerosis, and wherein the compound selected from the table 1 ab and Table 1c increases the fibrous cap thickness of the plaque.
  • the senescent cells are senescent preadipocytes, senescent endothelial cells, senescent fibroblasts, senescent neurons, senescent epithelial cells, senescent mesenchymal cells, senescent smooth muscle cells, senescent macrophages, or senescent chondrocytes.
  • the compound selected from the table 1 ab and Table 1c kills at least 20% of the senescent cells and kills no more than 5% of non-senescent cells in an organ or tissue comprising the senescent cells associated with the senescence associated disease or disorder. In another specific embodiment, the compound selected from the table 1 ab and Table 1c kills at least 25% of the senescent cells in an organ or tissue comprising the senescent cells associated with the senescence associated disease or disorder.
  • a method for treating osteoarthritis in a subject comprising administering to the subject a therapeutically-effective amount of a small molecule compound selected from the table 1 ab and Table 1c that selectively kills senescent cells over non-senescent cells, wherein (a) the compound selected from the table 1 ab and Table 1c is administered in at least two treatment cycles wherein each treatment cycle independently comprises a treatment course of from 1 day to 3 months followed by a non- treatment interval, and wherein the non-treatment interval is at least two weeks; or (b) the compound selected from the table 1 ab and Table 1c is administered directly to the osteoarthritic joint.
  • the compound selected from the table 1 ab and Table 1c induces collagen Type II production in the osteoarthritic joint.
  • compound selected from the table 1 ab and Table 1c inhibits erosion of a proteoglycan layer in the osteoarthritic joint. In another specific embodiment, the compound selected from the table 1 ab and Table 1c inhibits erosion of a bone of the osteoarthritic joint.
  • a method for inducing production of collagen Type II comprising administering to a subject in need thereof a therapeutically-effective amount of a Compound selected from the table 1 ab and Table 1c , which selectively kills senescent cells over non- senescent cells, wherein (a) the compound selected from the table 1 ab and Table 1c is administered in at least two treatment cycles wherein each treatment cycle independently comprises a treatment course of from 1 day to 3 months followed by a non-treatment interval, wherein the non-treatment interval is at least two weeks; or (b) the compound selected from the table 1 ab and Table 1c is administered directly to the osteoarthritic joint.
  • the compound selected from the table 1 ab and Table 1c is administered intra-articularly In another specific embodiment, the compound selected from the table 1 ab and Table 1c is administered topically, transdermally, or intradermally. In another specific embodiment, the compound selected from the table 1 ab and Table 1c is administered as a bolus infusion. In another specific embodiment, the compound selected from the table 1 ab and Table 1c is combined with at least one pharmaceutical excipient to formulate a pharmaceutical composition that provides timed release of the Compound selected from the table 1 ab and Table 1c . In another specific embodiment, the compound selected from the table 1 ab and Table 1c inhibits erosion of a proteoglycan layer in the osteoarthritic joint.
  • the compound selected from the table 1 ab and Table 1c inhibits erosion of a bone of the osteoarthritic joint. In another specific embodiment, the compound selected from the table 1 ab and Table 1c kills at least 20% of the senescent cells and kills no more than 5% of non-senescent cells in the osteoarthritic joint. In another specific embodiment, the compound selected from the table 1 ab and Table 1c kills at least 25% of the senescent cells in the osteoarthritic joint.
  • a method for treating a senescence-associated pulmonary disease or disorder in a subject comprising administering to the subject a therapeutically effective amount of a small molecule compound selected from the table 1 ab and Table 1c that selectively kills senescent cells over non-senescent cells, wherein the compound selected from the table 1 ab and Table 1c is administered as a monotherapy in at least two treatment cycles wherein each treatment cycle independently comprises a treatment course of from 1 day to 3 months followed by a non-treatment interval wherein the non-treatment interval is at least 2 weeks.
  • a method for treating a senescence- associated pulmonary disease or disorder in a subject comprising administering to the subject a Compound selected from the table 1 ab and Table 1c , which compound selected from the table 1 ab and Table 1c is a small molecule compound that selectively kills senescent cells, wherein the compound selected from the table 1 ab and Table 1c is administered in at least two treatment cycles, each cycle comprising a treatment course and a non-treatment interval, and wherein the non-treatment interval is at least 2 months.
  • the senescence-associated pulmonary disease or disorder is pulmonary fibrosis.
  • pulmonary fibrosis is idiopathic pulmonary fibrosis.
  • the senescence-associated pulmonary disease or disorder is chronic obstructive pulmonary disease (COPD).
  • COPD chronic obstructive pulmonary disease
  • the senescence-associated pulmonary disease or disorder is selected from age-related loss of pulmonary function, cystic fibrosis, bronchiectasis, emphysema, and asthma.
  • the compound selected from the table 1 ab and Table 1c is administered directly to an affected pulmonary tissue that comprises the senescent cells.
  • the compound selected from the table 1 ab and Table 1c is administered by inhalation, intranasally, intratracheally, or by intubation
  • the compound selected from the table 1 ab and Table 1c is administered as a bolus infusion.
  • the compound selected from the table 1 ab and Table 1c is combined with at least one pharmaceutical excipient to formulate a pharmaceutical composition that provides timed release of the Compound selected from the table 1 ab and Table 1c .
  • the compound selected from the table 1 ab and Table 1c kills at least 20% of the senescent cells and kills no more than 5% of non-senescent cells in a lung of the subject.
  • the compound selected from the table 1 ab and Table 1c kills at least 25% of the senescent cells in a lung of the subject.
  • a method for treating a cardiovascular disease or disorder caused by or associated with arteriosclerosis in a subject comprising administering to the subject a therapeutically- effective amount of a small molecule compound selected from the table 1 ab and Table 1c that selectively kills senescent cells over non-senescent cells, wherein the compound selected from the table 1 ab and Table 1c is administered in at least two treatment cycles wherein each treatment cycle independently comprises a treatment course from 1 day to 3 months followed by a non-treatment interval, wherein the non-treatment interval is at least 2 weeks.
  • the subject has atherosclerosis, congestive heart failure, peripheral vascular disease, hypertension, or coronary artery disease.
  • the cardiovascular disease or disorder is atherosclerosis.
  • the compound selected from the table 1 ab and Table 1c increases the stability of atherosclerotic plaque.
  • the compound selected from the table 1 ab and Table 1c reduces the lipid content of an atherosclerotic plaque in a blood vessel of the subject.
  • the compound selected from the table 1 ab and Table 1c increases the fibrous cap thickness of the plaque.
  • the compound selected from the table 1 ab and Table 1c inhibits formation of atherosclerotic plaque in a blood vessel of the subject.
  • the likelihood of occurrence of myocardial infarction, angina, stroke, carotid thrombosis, or coronary thrombosis is reduced.
  • a method for increasing the stability of atherosclerotic plaque present in a blood vessel of a subject comprising administering to the subject a therapeutically-effective amount of a small molecule compound selected from the table 1 ab and Table 1c that selectively kills senescent cells over non-senescent cells, wherein the compound selected from the table 1 ab and Table 1c is administered in at least two treatment cycles wherein each treatment cycle independently comprises a treatment course of from 1 day to 3 months followed by a non-treatment interval, wherein the non-treatment interval is at least 2 weeks.
  • the subject has a cardiovascular disease selected from atherosclerosis, congestive heart failure, peripheral vascular disease, hypertension, or coronary artery disease.
  • the cardiovascular disease or disorder is atherosclerosis.
  • the compound selected from the table 1 ab and Table 1c reduces the lipid content of an atherosclerotic plaque in a blood vessel of the subject.
  • the compound selected from the table 1 ab and Table 1c increases the fibrous cap thickness of the plaque.
  • the compound selected from the table 1 ab and Table 1c inhibits formation of atherosclerotic plaque in a blood vessel of the subject.
  • the compound selected from the table 1 ab and Table 1c reduces the amount of atherosclerotic plaque in a blood vessel of the subject.
  • the compound selected from the table 1 ab and Table 1c is administered parenterally or orally.
  • the compound selected from the table 1 ab and Table 1c is administered directly to an artery that comprises the senescent cells. In another specific embodiment, the compound selected from the table 1 ab and Table 1c is administered as a bolus infusion. In another specific embodiment, the compound selected from the table 1 ab and Table 1c is combined with at least one pharmaceutical excipient to formulate a pharmaceutical composition that provides timed release of the Compound selected from the table 1 ab and Table 1c . In another specific embodiment, the compound selected from the table 1 ab and Table 1c kills at least 20% of the senescent cells and kills no more than 5% of non-senescent cells in an arteriosclerotic artery of the subject.
  • the compound selected from the table 1 ab and Table 1c kills at least 25% of the senescent cells in an arteriosclerotic artery of the subject.
  • the treatment course is no longer than one month or no longer than two months.
  • the treatment course is (a) no longer than 5 days, (b) no longer than 7 days, (c) no longer than 10 days, (d) no longer than 14 days, or (e) no longer than 21 days.
  • the compound selected from the table 1 ab and Table 1c is administered every 2 nd day or every 3 rd day of the treatment course.
  • the treatment course is one day, two days, three days, or four days.
  • the compound selected from the table 1 ab and Table 1c is administered daily during the treatment course.
  • the non-treatment interval is (a) at least one month, (b) at least 2 months, (c) at least 3 months, (d) at least 6 months, (e) at least 9 months, or (f) at least 1 year.
  • the treatment course is one day and the non-treatment interval is between 0.5-12 months.
  • the treatment course is at least 5 days.
  • the compound selected from the table 1 ab and Table 1c is administered as a monotherapy.
  • the compound selected from the table 1 ab and Table 1c is administered in three or more treatment cycles.
  • each treatment course is no longer than one month or no longer than two months.
  • each treatment course is (a) no longer than 5 days, is (b) no longer than 7 days, is (c) no longer than 10 days, is (d) no longer than 14 days, or is (e) no longer than 21 days.
  • the compound selected from the table 1 ab and Table 1c is administered every 2 nd day or every 3 rd day of the treatment course.
  • each treatment course is one day, two days, three days, or four days.
  • the compound selected from the table 1 ab and Table 1c is administered daily during the treatment course.
  • the non-treatment interval is at least two weeks, one month, at least 2 months, at least 6 months, at least 9 months, or at least 1 year.
  • the compound selected from the table 1 ab and Table 1c to the subject comprises three or more treatment cycles.
  • the compound selected from the table 1 ab and Table 1c is administered as a monotherapy.
  • the senescence-associated disease or disorder is a cardiovascular disease selected from atherosclerosis, angina, arrhythmia, cardiomyopathy, congestive heart failure, coronary artery disease, carotid artery disease, endocarditis, coronary thrombosis, myocardial infarction, hypertension, aortic aneurysm, cardiac diastolic dysfunction, hypercholesterolemia, hyperlipidemia, mitral valve prolapsed, peripheral vascular disease, cardiac stress resistance, cardiac fibrosis, brain aneurysm, and stroke.
  • the subject has a cardiovascular disease selected from atherosclerosis, congestive heart failure, peripheral vascular disease, hypertension, or coronary artery disease.
  • the senescence-associated disease or disorder is inflammatory or autoimmune disease or disorder selected from osteoarthritis, osteoporosis, oral mucositis, inflammatory bowel disease, kyphosis, and herniated intervertebral disc
  • the senescence-associated disease or disorder is a neurodegenerative disease selected from Alzheimer's disease, Parkinson's disease, Huntington's disease, dementia, mild cognitive impairment, and motor neuron dysfunction.
  • the senescence-associated disease or disorder is a metabolic disease selected from diabetes, diabetic ulcer, metabolic syndrome, and obesity.
  • the senescence-associated disease or disorder is a pulmonary disease selected from idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, asthma, cystic fibrosis, emphysema, bronchiectasis, and age-related loss of pulmonary function.
  • the senescence-associated disease or disorder is an eye disease or disorder selected from macular degeneration, glaucoma, cataracts, and vision loss.
  • the senescence-associated disease or disorder is an age-related disorder selected from renal disease, renal failure, frailty, hearing loss, muscle fatigue, skin conditions, skin wound healing, liver fibrosis, pancreatic fibrosis, oral submucosa fibrosis, and sarcopenia.
  • the senescence-associated disease or disorder is a dermatological disease or disorder is selected from eczema, psoriasis, hyperpigmentation, nevi, rashes, atopic dermatitis, urticaria, diseases and disorders related to photosensitivity or photoaging, rhytides; pruritis; dysesthesia; eczematous eruptions; eosinophilic dermatosis; reactive neutrophilic dermatosis; pemphigus; pemphigoid; immunobullous dermatosis; fibrohistocytic proliferations of skin; cutaneous lymphomas; and cutaneous lupus.
  • the senescence-associated disease or disorder is atherosclerosis; osteoarthritis; idiopathic pulmonary fibrosis; or chronic obstructive pulmonary disease.
  • n the senescence-associated disease or disorder is osteoarthritis and the compound selected from the table 1 ab and Table 1c is administered directly to the osteoarthritic joint.
  • the compound selected from the table 1 ab and Table 1c is administered intra-articularly to the osteroarthritic joint.
  • the compound selected from the table 1 ab and Table 1c is administered topically, transdermally, or intradermally.
  • the senescence-associated disease or disorder is osteoarthritis and the compound selected from the table 1 ab and Table 1c induces production of Type II collagen in a joint.
  • the senescence-associated disease or disorder is osteoarthritis and the compound selected from the table 1 ab and Table 1c inhibits erosion of a proteoglycan layer in a joint.
  • the senescence-associated disease or disorder is osteoarthritis and the compound selected from the table 1 ab and Table 1c inhibits erosion of a bone of a joint.
  • the senescence-associated disease or disorder is idiopathic pulmonary fibrosis and the compound selected from the table 1 ab and Table 1c reduces the amount of fibrotic pulmonary tissue in the lung.
  • the compound selected from the table 1 ab and Table 1c is administered intranasally, by inhalation, intratracheally, or by intubation.
  • the compound selected from the table 1 ab and Table 1c is combined with at least one pharmaceutically acceptable excipient to formulate a pharmaceutically acceptable composition to provide timed-release of the Compound selected from the table 1 ab and Table 1c .
  • the compound selected from the table 1 ab and Table 1c is administered as a bolus infusion.
  • the senescence-associated disease or disorder is atherosclerosis, and wherein the compound selected from the table 1 ab and Table 1c increases stability of atherosclerotic plaque.
  • the senescence-associated disease or disorder is atherosclerosis, and wherein the compound selected from the table 1 ab and Table 1c inhibits formation of atherosclerotic plaque in a blood vessel of the subject.
  • the senescence-associated disease or disorder is atherosclerosis, and wherein the compound selected from the table 1 ab and Table 1c reduces the lipid content of an atherosclerotic plaque in a blood vessel of the subject.
  • the senescence-associated disease or disorder is atherosclerosis, and wherein the compound selected from the table 1 ab and Table 1c increases the fibrous cap thickness of the plaque.
  • the senescence-associated disease or disorder is atherosclerosis, and wherein the likelihood of occurrence of myocardial infarction, angina, stroke, carotid thrombosis, or coronary thrombosis is reduced.
  • the senescent cells are senescent preadipocytes, senescent endothelial cells, senescent fibroblasts, senescent neurons, senescent epithelial cells, senescent mesenchymal cells, senescent smooth muscle cells, senescent macrophages, or senescent chondrocytes.
  • the compound selected from the table 1 ab and Table 1c kills at least 20% of the senescent cells and kills no more than 5% of non-senescent cells. In another specific embodiment, the compound selected from the table 1 ab and Table 1c kills at least 25% of the senescent cells.
  • the compound selected from the table 1 ab and Table 1c is used interchangeably with the therapeutic agent reducing, inhibiting or degrading at least one of the Targets.
  • the therapeutic agent reducing, inhibiting or degrading at least one of the Targets is a small molecule.
  • the therapeutic agent reducing, inhibiting or degrading at least one of the Targets is a gene therapy. The exlpanation behind the selection of compounds listed in Table 1ab, Table c and Table d. Table 1a 1.
  • DNN deep neural network
  • Therapeutic target classifications for 1005 drugs can be obtained from the Anatomical Therapeutic Chemical Classification System (ATC) provided by DrugCentral. There are 14, 79, 182, and 444 classes for ATC levels 1, 2, 3, and 4, respectively.
  • Biological protein target annotations for 135 target classes and 411 drugs can be obtained from ChEMBL.
  • ECFPs extended- connectivity fingerprints
  • MACCS Molecular ACCess System
  • Figure 1D Embedding network architecture and training loss for a single expression profile.
  • Inputs are standardized L1000 profiles and are processed by a densely connected neural network.
  • the output embeddings are used to predict the class (perturbagen identity) of the input by softmax where logits are cosine similarities between the profile embeddings and learned class embeddings, scaled by a learned constant.
  • the prediction cross-entropy loss is used to train the network.
  • the network is trained with a modified softmax cross-entropy loss over n classes, where classes are perturbagen identities. For ease of notation, the following describes the loss for a single sample. The loss for an entire batch is defined similarly.
  • the margin m are linearly increased at a rate of 0.0002 per step up to a maximum 0.25. These values are chosen without a hyperparameter search and are therefore unlikely to be optimal.
  • the denoised representation was used as it is described in the attached Article 1 (“Integration of genetic, transcriptomic, and clinical data reveals pharmacological modulators of human longevity”) to obtain the risk model from human genetics data which can be translated to the L1000 perturbagens expression profiles.
  • the selected targets are: ABCB1, ADRA1A, ADRA1B, ADRA1D, BRD2, BRD3, BRD4, DRD2, DRD3, DRD4, DRD5, EBP, EGFR, EPHA6, EPHA8, ERBB2, HDAC1, HDAC10, HDAC11, HDAC2, HDAC3, HDAC4, HDAC5, HDAC6, HDAC7, HDAC8, HDAC9, HRH1, HSP90AA1, HTR2A, HTR2B, HTR2C, HTR5B, HTR6, HTR7, KCNH2, MAP4K5, MTOR, PIK3C2A, PIK3C2B, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PIK3R1, PRKDC, SLC6A2, SLC6A3, SLC6A4 (List of Targets 1a).
  • RNA-seq experimental proprietary dataset consisting of four mutant groups (daf-2(e1370), age-1(mg44) [at the first and second gen- erations of homozygosity], and daf-2(e1391); daf-12(m20) double mutant), the three examples of life- extending RNAi (daf-4, che-3 and cyc-1, representing knockdown of diverse pathways) and two independent control runs represented by C. elegans wild-type (Bristol-N2, strain DRM); 60 transcriptomes in total; mean adult lifespan extended 2.2- to 9.4-fold. 2.
  • transcriptomic aging signature [1] the list of genes that have statistically significant association with age rescaled by lifespan (the first list). 3. Genes in the aging signature that have positive correlation with age rescaled by lifespan are over-expressed in late life of the animals, and if a gene’s correlation is negative its gene expression decreases with age. 4. Since long-lived mutant lines had 9.4-fold increased lifespan with respect to the control strains and showed the time scaling of gene expression changes — the transcriptome of long- lived mutant strains changed similar to the control strains but proportionally slower according to their lifespan. 5. The identified aging signature represents a vector of the aging-related changes of gene expression.
  • the PI3K-null transcriptomic signature For extracting the targets of the second list, (the PI3K-null transcriptomic signature), we corrected for age-related changes in all animals by subtracting aging-related changes in each of the strains. This was done by projecting out all gene expression changes along the aging signature. 6. After that, by gene-wise linear regression, we identified the list of genes that had statistically significant correlation with lifespan of long-lived strains. That list represents the PI3K-null signature. Genes in the PI3K-null signature that have positive correlation with lifespan are over-expressed in long-lived strains, and vise versa, genes with negative correlations have lower gene expression levels in long-lived mutants with respect to the control lines. 7.
  • Targets 1b CDK7 , CHEK2 , CAPN1 , CTSB , HSP90AB1 , HSP90AA1 , HSPA5 , NFKB1 , NPC1 , PABPC1 , ABCB11 , ABCB1 , PLK1 , PLK3 , ADRA2A , ADRA2C , ADRA2B , TUBB6 , SENP8 , SENP6 , SENP7 , MMP14 , MMP13 , MMP8 , MMP3 , MMP1 , MAPKAPK2 , CTSL , CTSK , CTSS , ATXN2 , TBXAS1 , CREBBP , PDGFRA , FLT4 , FLT1 , KDR , PDGFRB ,
  • Figure F Survivals of C.elegans strains treated with the 8 compounds selected for the lifespan screening assay.
  • Table T1a The output of CMAP tool [16] for the given PI3K-null transcriptomic signature. Score is the CMAP score for similarity of a drug’s profile to the PI3K-null transcriptomic signature. Name is the drug name. Description is the mechanism of action of the given drug.
  • Table T1b The same as Table T1a, but obtained with the older version of CMAP [2,3].
  • coli HT115 expressing double-stranded RNAs for target-gene knockdowns [5] for both RNA- preparation and lifespan studies. Survivals Lifespan assays were conducted at 20 °C, as described previously [5]. Briefly, synchronous cultures were initiated by lysis of gravid hermaphrodites in alkaline hypochlorite. Worms were selected at the L4 larval stage, placed 50 worms per plate, and transferred at 1- to 2-day intervals onto fresh plates during days 1–7, and at 2- to 3- day intervals after that. A worm was scored as dead if it failed to move, either spontaneously or in response to a mechanical stimulus; lost worms were excluded (censored) from the survival analysis.
  • Worms were washed off plates and rinsed twice in survival buffer; after 30 min at 20 °C (to allow digestion of enteral bacteria), they were flash frozen and stored at ⁇ 80 °C.
  • Frozen worms were ground in a dry-ice-cooled mortar and pestle, and total RNA was extracted using RNeasy RNA extraction kits (Qiagen), followed by RNA purification for construction of transcript libraries using TruSeq RNA kits (Illumina, v.2). Sequences are generated as PE100 multiplexes, 100-bp paired-end reads from an Illumina HiSeq2500 or NextSeq instrument, producing 40–50 ⁇ 106 reads per sample.
  • RNA-seq dataset RNA-seq reads were mapped to the C. elegans genome (WBcel235, Ensembl annotation) using TopHat 2.1.1 (with–b2-very-sensitive and–GTF options) [7] and gene- level read counts were obtained using the htseq-count software [8].
  • Low-expressed genes with at least one zero read count per sample were removed from subsequent analysis.
  • Raw read counts were normalized using the upper quartile method [9] and converted to RPKM values using the edgeR library [10].
  • CMAP Connectivity Map
  • OrthoList database [11] comprising information from four other databases: Ensembl Compara [12], InParanoid [13], NCBI HomoloGene Database, OrthoMCL [14]. Since, CMAP requires human genes to be presented by HG-U133A tags (Affymetrix Human Genome U133A Array), the g:Profiler database [15] was used to map human Ensembl gene IDs to HG-U133A tags.
  • Table 1ab consists of all compounds from the Table 1a and all compounds from the Table 1b created as shown above and the just one of the each repetitious compounds is left in the table 1ab
  • We also added the wording about analogs of compounds from TABLE 1c will have anti- aging properties.
  • GWAS Genome-Wide Association Studies
  • WES whole-exome sequencing
  • UK Biobank UK Biobank
  • LEF loss of function
  • a survey of LOF variants in humans predicted 100 LOF variants per individual, including 20 genes that carry homozygous LOFs and hence are likely to be non-functional [7].
  • RESULTS Deep neural network for batch removal in the LINCS L1000 dataset The LINCS L1000 dataset is a unique resource for associating genetic and pharmacological perturbations with gene expression patterns, which is a key feature of our current study. However, the dataset suffers from non-biological variance effects and the need for dimensionality reduction.
  • DNN deep neural network
  • the network was trained using over 1.2M cell culture samples from ⁇ 80 cell lines and all molecular and genetic perturbations including more than 7 measurements each.
  • consensus profiles (signatures) treating different doses of the same molecular perturbation as separate biological conditions.
  • the procedure involved averaging over all available repeats including measurements of the same perturbation in different cell lines and times points.
  • Most of the transcriptomic signatures of small molecule perturbations in LINCS L1000 were only available at a very high 10 ⁇ M concentration and hence, for consistency, only the measurements at this concentration were used in the calculations below.
  • the dots stand for other covariates, such as regional affiliations/assessment centers, ethnic variables, and principal components (PCs) of UKB genotypes [17].
  • the coefficients ⁇ , ⁇ , and ⁇ g are the Cox regression coefficients characterizing the effects of age, sex, and loss of a gene g.
  • the direct solution of the full regression problem (1) is unfeasible.
  • To produce a tractable and numerically stable solution we followed the two-step procedure.
  • the parameters of the first-order death risk model are shown in Fig.2.
  • the log-linear regression coefficient for the chronological age in the lifespan model was 0.106 per year, which is consistent with a typical estimate of the Gompertz slope parameter in the range of 0.06 ⁇ 0.11 per year [18].
  • the model produced increased mortality estimates associated with smoking, Townsend deprivation index, and increased BMI.
  • the pathway activation scores are representations of the transcriptomic shifts due to the germline PTV mutations and can be used as additional covariates in the multivariate proportional hazards model.
  • the transcriptomic shifts PA n can be used instead o n ⁇ f the genetic profiles PTV g in the Cox- proportional hazard model in Eq.1.
  • the genetic component (the linear combination of the pathway activation scores, of the risk model is hereinafter referred to as “the genetic longevity score” or GLS.
  • the sign convention is such that larger GLS values correspond to the lower risk and hence predict longevity.
  • gene transcripts associated with the longevity phenotype For this, we used full transcriptomes from LINCS (12K gene transcript each) and ran the correlation between gene transcript levels and the genetic longevity score computed for the same samples using the gene expression profile embeddings.
  • the DNN can translate any L1000 sample into PA scores, which, in turn, can be used to compute the longevity score, corresponding to the negative risk associated with the drugs.
  • the values can be used to rank- order drugs or genetic interventions according to the expected life-extending effect (the higher expected effect is associated with the larger value of the score).
  • LOPAC Library of Pharmacologically Active Compounds
  • the inferred toxicity score that is the log-odds ratio of the toxicity prediction model, could be computed for every molecular perturbation from LINCS L1000 and exhibited no meaningful correlation with GLS
  • the deep neural network architecture employed here provides a superior performance relative to the state-of-the-art batch removal techniques.
  • the publicly available Fc1000 dataset also provides the transformed representations of shRNA and drug perturbations and, therefore, can be used to train the all-cause mortality model from UKB data.
  • Neither of the models trained from the raw L1000 or Fc1000 transformed datasets were able to produce a significant enrichment from the list of life-extending and -reducing compounds from [12] using the direct compound scores (Figs.10 and 12, respectively).
  • Target-deconvolution universally improved the results (Figs.11 and 13, respectively), and calculations in the raw and Fc1000 datasets led to significant enrichment of life-extending compounds from the experiment.
  • the Fc1000-based analysis produced less enrichment (ROC AUC of 0.63 vs.0.72, see Figs.13 and 6, respectively) and returned fewer (30 vs.36, not shown) targets associated with human longevity than our model.
  • the Fc1000-based GLS score also negatively and significantly enriched the list of drugs reducing the lifespan of nematodes in the experiment.
  • DISCUSSION GWAS has been criticized for the interpretability of its findings and the small effect sizes of most risk variants.
  • mice Most of the current mechanistic insights into the basic biology of aging originate from studies of lab animals, such as nematodes [], fruit flies [] or mice []. Mice is currently the predominant preclinical model of aging and age-related diseases, but it is not universally applicable, e.g. mice are not champions of longevity and, unlike humans, they mostly die of cancer [ref]. Therefore, it is an open question as to what the best strategy is when it comes to the identification of anti-aging interventions for successful therapeutic application in humans. GWAS of human lifespan, including examining its proxies, such as extreme lifespan, parental survival, and healthspan, produced a number of gene variants potentially associated with human aging.
  • GWAS on centenarians consistently associate loci near APOE gene with extreme longevity, and, depending on population, loci near FOXO3A, HLA-DQA1 and SH2B3 ⁇ cite ⁇ melzer2019 ⁇ .
  • GWAS for several longevity proxies such as parental lifespan ⁇ cite ⁇ Pilling2017 ⁇ and healthspan ⁇ cite ⁇ zenin2018identification ⁇ , confirmed most of the longevity variants and identified additional ones.
  • large collections of transcriptomes such as LINCS and L1000, or its earlier version CMAP, have been used extensively for in silico drug repurposing in general and specifically in aging ⁇ cite ⁇ .
  • the genetic longevity score, GLS is a combined effect of a multitude of ultra-rare naturally occurring germline mutations.
  • LOF in BRCA2 or ASXl1 in this study
  • HR ⁇ 1.1 the integration of LOF on a pathway level exemplified in this study led to the additional explained variance (0.5 years) of lifespan on par with other established approaches.
  • the GLS can be computed for every perturbation and works very well in differentiating between the life-extending and shortening compounds in the experiment ⁇ cite ⁇ ye2013pharmacological ⁇ (Fig. ⁇ ref ⁇ fig:ci_genetic_ls ⁇ ).
  • Fig. ⁇ ref ⁇ fig:ci_genetic_ls ⁇ There could be two explanations of these observations: a technical one and the one of more principal importance.
  • to ⁇ cite ⁇ ye2013pharmacological ⁇ the life-extending compounds in the experiment were measured twice, in the primary and confirmatory screens. In contrast, the compounds reducing lifespan were tested only once. Therefore, the list of life-extending compounds may be more reliable in principle.
  • Target deconvolution may be more reliable but has the obvious drawback.
  • the procedure may only work for the drug targets (MOAs) with a sufficient number of measurements in LINCS L1000 and ChEMBL.
  • MOAs drug targets
  • the choice of LOPAC library in the experiment also suffers from a similar bias: pharmacologically active compounds reflect GPCR modulators' over-representation, comprising XXX ⁇ % of drugs sold in pharmacies.
  • the gene expression levels measured in LINCS L1000 are dominated by batch effects.
  • HDACs histone deacetylases
  • the drugs revert the age-related alteration in histone acetylation that in turn activated expression of pro-longevity transcription factors in mice models of age-related diseases ⁇ cite ⁇ mcintyre2019molecular ⁇ .
  • Pro-longevity effects of HDAC inhibitors in model organisms are now supported by our analysis of human genetics. Since the drugs are already used in neurology and cancer treatments, we propose a further clinical investigation of the HDAC inhibitors against aging and frailty. A significant number of top-scoring drugs and targets are linked to serotonin and dopamine signaling.
  • serotonin modifying drugs are widely used in psychiatry and neurology, the effect of serotonin is not limited to CNS, it regulates numerous biological processes outside the brain including cardiovascular, gastrointestinal and endocrine function ⁇ cite ⁇ berger2009expanded ⁇ . There are conflicting results regarding the life-extending effect of this group of drugs.
  • melanogaster ⁇ cite ⁇ ro2016serotonin ⁇ by altering protein food value perception it did not affect lifespan in fixed-diet conditions and increased lifespan of flies that were allowed to chose nutrients. It would be harder to expect that such complicated biology may be reproduced in cell- based experiments in LINCS L1000, therefore the appearance of pharmacological targets involved in serotonin and dopamine signalling may be an artifact of our computational procedure. Even if the predictions of our network are real, we believe that the longevity effects of the drugs from this class would be hard to confirm and study in animal models and clinical trials.
  • the LINCS L1000 dataset provides transcriptional responses of multiple human cells to genetic interventions such as shRNA ($4000$ knockouts out of roughly $20k$ protein-coding genes in the human genome ⁇ cite ⁇ salzberg2018open ⁇ ).
  • shRNA ($4000$ knockouts out of roughly $20k$ protein-coding genes in the human genome ⁇ cite ⁇ salzberg2018open ⁇ .
  • the dimensionality reduction achieved by the deep neural network allowed to build and cross-validate the all-cause mortality risk model in UKB with as little as $ ⁇ 200k$ exomes available.
  • the DNN was constructed of the three blocks: DenseNet block, Embeddings, and Perturbagen classifier, see Fig. ??.
  • the DenseNet was implemented as suggested in [13] with 32 hidden layers and a growth rate of 48.
  • the Embeddings layer is a dense layer with 2514 Input size and 20 output size.
  • the perturbagen classifier was used to predict the perturbagen class (pert_iname label in the L1000 dataset) from the embedding vector using additive margin Softmax (AM-Softmax) [22] with the margin value was set to 0.2 as in [13].
  • AM-Softmax additive margin Softmax
  • the training data included profiles from LINCS PHASE I (GEO accession number GSE92742) and PHASE II (GEO accession number GSE70138).
  • the preprocessed dataset involving 1467244 gene expression profiles corresponding to 27870 unique compound perturbation classes was split into the training and test datasets at 80/20 ratio.
  • the test dataset did not include gene expression signatures from the training dataset.
  • Risk models with small effects MRR log-likelihood test cross-validation Gene set enrichment analysis To obtain insights into the biology of human longevity we used LINCS L1000 dataset for search of gene transcripts most associated with the longevity risk scores.
  • FIG.1 Schematic representation of the development and characterization of the AI/ML system for integrative analysis of big clinical, transcriptomic, and genetic data.
  • A) Biological insights are obtained by capturing clinically relevant rare loss of function mutations (LOF) in the UKB repository of medical history and WES data of 200k subjects.
  • B) The effects of LOF mutations are aggregated on the pathway level with the help of a Deep Neural Network (DNN) producing batch removal and dimensionality reduction (embedding).
  • DNN Deep Neural Network
  • FIG.3 GO Biological Process (a) and KEGG pathway (b) enrichment of gene transcripts associated with the genetic longevity score (GLS).
  • FIG.4 Gene sets associated with diseases from GLAD4U database and over-represented among gene transcripts associated with GLS.
  • FIG.5 Drugs scored by genetics risk predicts life-extending drugs in C. elegans screening [12]
  • FIG.6 Targets recovered from lifespan signature and ChEMBL database
  • FIG.7 LoF burden is associated with survival in UKB model.
  • the significance of the association in the Cox proportional hazards model increases with the number of samples in analysis at any level of MAF. The best effect is found for ultra rare variants MAF ⁇ 0.0001.
  • FIG.8 The significance of the PTV/LOF types association with survival in full UKB dataset.
  • FIG.9 The Manhattan plot summarizing the associations of individual LOF (at MAF ⁇ 1e ⁇ 3 with survival in the full dataset of 200k UKB genotypes.
  • FIG.10 Drugs scored by genetics risk predicts life-extending drugs in C. elegans screening [12], predicted on raw data 5 157
  • FIG.11 Targets recovered from lifespan signature and STITCH database , predicted on raw data
  • FIG.12 Drugs scored by genetics risk predicts life-extending drugs in C. elegans screening [12], predicted on FC1000 data
  • FIG.13 Targets recovered from lifespan signature and STITCH database predicted on FC1000 data
  • All Examples 1-35 are prophetic Example 1 Animal treatment/In vivo experiments
  • the animals are treated with the compound selected from the table 1 ab and Table 1c or composition, comprising such compound via IV injection.
  • the animals are treated with the compound selected from the table 1 ab, Table 1c and Table 1d, including but not limited to compounds selected from the group consisting of: CYCLOSPORINE , TARIQUIDAR , MARIMASTAT , PRINOMASTAT , APRATASTAT , YOHIMBINE , QUETIAPINE , DOXEPIN , MIANSERIN , ERGOTAMINE , PIPAMAZINE , PHENTOLAMINE , LISURIDE , TAMSULOSIN , DEXMEDETOMIDINE , INDORAMIN , RISPERIDONE , SERTINDOLE , XYLOMETAZOLINE , NAPHAZOLINE , TETRAHYDROZOLINE , DESIPRAMINE , ALFUZOSIN , SILODOSIN , TERAZOSIN , OLANZAPINE , ZOTEPINE , DROPERIDOL
  • the animals are treated subcutaneously.
  • the animals are treated by oral gavage.
  • the animals are treated orally with the food mixed with the compound selected from the table 1 ab and Table 1c .
  • Any one of the compounds selected from Tables 1 ab and Table 1c are dissolved in at least one of the following ways: 1) dissolved in DMSO to 10 mM stocks and used in the final assay buffer with minimum 1% DMSO (for 10 micro M final measurements) and with 2% DMSO (for 200 micro M final measurements), 2) dissolved in water to 200 micro M stock solution.
  • the stock solution is then used for further dilutions with assay buffer, and 3) any other dissolution known in the field.
  • the compound selected from the table 1 ab and Table 1c in any other example is dissolved by any one of the methods described if better method is not suggested here or by the expert.
  • the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a dosage 20 times less than its LD50.
  • the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a Maximum Tolerated dose.
  • the compound selected from the table 1 ab and Table 1c is administered by intraperitoneal injection (IP) in a doses: group #1 - 1 mg/kg, group #2 - 10 mg/kg, group #3 - 100 mg/kg.
  • IP intraperitoneal injection
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a dosage selected from the group consisting of: 10 times less than its LD50, 50 times less than its LD50, 100 times less than its LD50, 500 times less than its LD50, 1000 times less than its LD50, 5000 times less than its LD50, 10000 times less than its LD50, Maximum Tolerated dose, or in the dosage in which such compound was found effective in mice for at least one other indication known in the art.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in the one of the following regimens: once, once a day for 1 week, once every two days for 1 week, once every two days for 2 weeks, once every week for 1 month, once every two days for 1 month, once every two days or once every week until at least one symptom of aging is alleviated, in the regimen in which this compound was effective in animal model for its initial indication, in any other regimen described in this application, in any other regimen reasonably defined by the expert in the field taking into the account the knowledge about the compound.
  • compound selected from the table 1 ab and Table 1c is compound selected from the group consisting of: CYCLOSPORINE , TARIQUIDAR , MARIMASTAT , PRINOMASTAT , APRATASTAT , YOHIMBINE , QUETIAPINE , DOXEPIN , MIANSERIN , ERGOTAMINE , PIPAMAZINE , PHENTOLAMINE , LISURIDE , TAMSULOSIN , DEXMEDETOMIDINE , INDORAMIN , RISPERIDONE , SERTINDOLE , XYLOMETAZOLINE , NAPHAZOLINE , TETRAHYDROZOLINE , DESIPRAMINE , ALFUZOSIN , SILODOSIN , TERAZOSIN , OLANZAPINE , ZOTEPINE , DROPERIDOL , THIORIDAZINE , CHLORPROMAZINE , FLUPHENAZINE , CAR
  • the anti-aging effect compound selected from the table 1 ab and Table 1c is administered by the root of administration, in dosage and regimen which were previously used (known in the art ) to show the efficacy in mice (or if mice model had not been done in any other animal model, but with the correction in the dosage for the mice) of the same compound in at least one disease or condition.
  • the dosage is taken two-fold greater than the dosage in which the compound was effective for other disease in mice, or in yet another example the dosage is taken two- fold less.
  • the dosage is taken ten-fold greater than the dosage in which the compound was effective for other disease in mice, or in other example the dosage is taken ten-fold less than the dosage in which the compound was effective for other disease in mice.
  • the mice from reference group are treated with 42 ppm of a well-characterized life- extending drug - rapamycin as the positive control. The life-extending properties of rapamycin have been confirmed in multiple experiments, including a large study within the NIH (NIA) Intervention Testing Program (https://www.nia.nih.gov/research/dab/interventions-testing-program-itp).
  • the interventions in the animals can reduce their age related morbidity and mortality.
  • the Frailty Index is measured as in Whitehead et al (2014) and consists of 31 phenotypes that are indicators of age-associated health deterioration including alopecia, physical/musculoskeletal, auditory, ocular, nasal, signatures of digestive and other disorders. Each phenotype is scored on a 0, 0.5 or 1 scale, based on its severity.
  • the 31 metrics share many characteristics of the human frailty indicators and were previously reported to progress similarly with aging in mice and humans (Kane, A.E., et al, 2016).
  • Synaptoplasticity Progressive reduction of structural and functional plasticity is associated with the gradual decline in cognitive function. Moreover, chemotherapy-induced cognitive decline is associated with loss of synaptic integrity and it was demonstrated that cisplatin-treated mice show reduced levels of PSD95 and synaptophysin (Chiang ACA et al., 2019). Synaptophysin immunoreactivity has been identified as a useful marker for presynaptic density, whereas PSD95 is a marker of post-synaptic density. In order to analyze synaptic plasticity in the experiment, Western blot analysis of mouse brain tissue are performed using antibodies against PSD95 and synaptophysin.
  • mice (10 weeks) are fed a high fat diet (HFD) (Harlan Teklad TD.88137) having 42% calories from fat, beginning at Week 0 and throughout the study.
  • HFD high fat diet
  • mice (10 weeks) are fed normal chow (- HFD).
  • - HFD normal chow
  • From weeks 0-2, one group of HFD mice and -HFD mice are treated with compound selected from the table 1 ab and Table 1c with the dosage and root of administration selected from the Example 1.
  • the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a dosage 20 times less than its LD50 by intraperitoneal injection.
  • the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a Maximum Tolerated dose by intraperitoneal injection.
  • the compound selected from the table 1 ab and Table 1c is administered by intraperitoneal injection (IP) in a doses: group #1 - 1 mg/kg, group #2 - 10 mg/kg, group #3 - 100 mg/kg.
  • IP intraperitoneal injection
  • the treatment by the compound selected from the table 1 ab and Table 1c consists in single administration of the compound selected from the table 1 ab and Table 1c in the dosage selected from this example.
  • dosage, regimen and root of administration are selected from the Example 1.
  • one treatment cycle is 14 days treatment, 14 days off.
  • Vehicle is administered to one group of HFD mice and one group of -HFD mice.
  • week 4 timepoint 1
  • one group of mice are sacrificed and to assess presence of senescent cells in the plaques.
  • the compound selected from the table 1 ab and Table 1c and vehicle administration is repeated from weeks 4- 6.
  • week 8 timepoint 2
  • the mice are sacrificed and to assess presence of senescent cells in the plaques.
  • the remaining mice are treated with the compound selected from the table 1 ab and Table 1c or vehicle from weeks 8-10.
  • week 12 timepoint 3
  • the mice are sacrificed and to assess the level of plaque and the number of senescent cells in the plaques.
  • Values are normalized to GAPDH and expressed as fold-change versus age-matched, vehicle- treated LDLR "7" mice on a normal diet.
  • the data show that clearance of senescent cells with the compound selected from the table 1 ab and Table 1c in LDLR "7" mice fed a HFD reduce expression of several SASP factors and senescent cell markers, MMP3, MMP13, PAI1, p21, IGFBP2, IL-1A, and IL-1B after 1 treatment cycle.
  • LDLR "7" mice fed a HFD and treated with the compound selected from the table 1 ab and Table 1c A or vehicle are sacrificed, and aortas are dissected and stained with Sudan IV to detect the presence of lipid.
  • Body composition of the mice are analyzed by MRI, and circulating blood cells are counted by Hemavet.
  • the data show that treatment with the compound selected from the table 1 ab and Table 1c reduces plaques in the descending aorta by -45%( The platelet and lymphocyte counts are equivalent between the compound selected from the table 1 ab and Table 1c and vehicle treated mice.
  • the treatment with the compound selected from the table 1 ab and Table 1c also decreases mass and body fat composition in mice fed a HFD.
  • STUDY The study assesses the extent to which the compound selected from the table 1 ab and Table 1c based clearance of senescent cells from LDLR "7" /3MR double transgenic mice improves pre-existing atherogenic disease.
  • LDLR " 73MR double transgenic mice (10 weeks) and LDLR "7" single transgenic mice (10 weeks) are fed a high fat diet beginning at Week 0 until Week 12.
  • the compound selected from the table 1 ab and Table 1c is administered to groups of mice (dosages for respective groups: 0.1 mg/kg – for group #1, 1 mg/kg – for group #2, 10mg/kg – for group #3, 100mg/kg – for group #4 and 1g/kg – for group #5) intraperitoneally from weeks 12-13 and weeks 14-15.
  • the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a dosage 20 times less than its LD50 by intraperitoneal injection.
  • the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a Maximum Tolerated dose by intraperitoneal injection.
  • the compound selected from the table 1 ab and Table 1c is administered by intraperitoneal injection (IP) in a doses: group #1 - 1 mg/kg, group #2 - 10 mg/kg, group #3 - 100 mg/kg.
  • IP intraperitoneal injection
  • the treatment by the compound selected from the table 1 ab and Table 1c consists in single administration of the compound selected from the table 1 ab and Table 1c in the dosage selected from this example.
  • dosage, regimen and root of administration are selected from the Example 1. At week 16, the level of plaque and the number of senescent cells in the plaques are determined.
  • Example 3 AN ANIMAL STUDY FOR EVALUATING OF THE SENOLYTIC EFFECT OF COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c IN MICE
  • Senescence in animals can be induced through the administration of doxorubicin followed by treatment of the compound selected from the Table 1 ab and Table 1c .
  • mice are sacrificed, and fat and skin are collected for RNA analysis, while lungs are collected and flash frozen for immunomicroscopy analysis.
  • RNA is analyzed for expression of SASP factors (mmp3, IL-6) and senescence markers (p21, i 6, and p53). Frozen lung tissue is analyzed for DNA damage marker ( ⁇ 2 ⁇ ).
  • the mice to be tested contain a transgene insertion of p16-3MR.
  • 3MR tri-modality reporter
  • LOC Renilla luciferase
  • mRFP monomeric red fluorescence protein
  • tTK truncated herpes simplex virus
  • GMV ganciclovir
  • the 3 MR cDNA is inserted in frame with p16 in exon 2, creating a fusion protein containing the first 62 amino acids of pi 6, but does not include the full- length wild-type p16 protein. Insertion of the 3MR cDNA also introduces a stop codon in the p 19 ARF reading frame in exon 2.
  • the effect of the compound selected from the Table 1 ab and Table 1c is analyzed by the reduction of luminescence intensity.
  • Female C57/B16 p16-3MR mice are treated with Doxorubicin. Luminescence is measured 10 days later and used as baseline for each mouse (100% intensity).
  • the compound selected from the table 1 ab and Table 1c is administered to groups of mice (dosages for respective groups: 0.1 mg/kg – for group #1, 1 mg/kg – for group #2, 10mg/kg – for group #3, 100mg/kg – for group #4 and 1g/kg – for group #5) intraperitoneally daily from day 10 to day 24 post- doxorubicin treatment.
  • the compound selected from the Table 1 ab and Table 1c is administered according to the protocol selected from the Example 1.
  • the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a dosage 20 times less than its LD50 by intraperitoneal injection daily from day 10 to day 24 post- doxorubicin treatment.
  • the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a Maximum Tolerated dose by intraperitoneal injection daily from day 10 to day 24 post- doxorubicin treatment.
  • the compound selected from the table 1 ab and Table 1c is administered by intraperitoneal injection (IP) in a doses: group #1 - 1 mg/kg, group #2 - 10 mg/kg, group #3 - 100 mg/kg.
  • IP intraperitoneal injection
  • the treatment by the compound selected from the table 1 ab and Table 1c consists in single administration of the compound selected from the table 1 ab and Table 1c in the dosage selected from this example.
  • dosage, regimen and root of administration are selected from the Example 1. Luminescence is then measured at day 7, 14, 21, 28, 35 post- compound selected from the Table 1 ab and Table 1c treatments, and final values calculated as % of the baseline values. Control animals (DOXO) are injected with equal volume of PBS.
  • the level of mRNA of endogenous mmp-3, IL-6, p21, p16, and p53 in the skin and fat from animals after treatment with doxorubicin alone (DOXO) or doxorubicin plus compound selected from the Table 1 ab and Table 1c is plotted. The values represent the fold induction of the particular mRNA compared with untreated control animals.
  • Immunofluorescence microscopy of lung sections from doxorubicin treated animals (DOXO) and doxorubicin and compound selected from the Table 1 ab and Table 1c can be detected by binding to a primary rabbit polyclonal antibody specific for ⁇ 2 ⁇ followed by incubation with a secondary goat anti- rabbit antibody, and then counterstained with DAPI.
  • the percent positive cells from immunofluorescence microscopy are calculated and can be represented as percentage of the total number of cells.
  • Data can be obtained from doxorubicin-treated mice (Doxo), and doxorubicin + compound selected from the Table 1 ab and Table 1c -treated mice).
  • the compound selected from the Table 1 ab and Table 1c can be analyzed for reduced senescence- associated (SA) ⁇ - galactosidase ( ⁇ -gal) intensity of fat biopsies from animals first treated with doxorubicin.
  • SA reduced senescence- associated
  • ⁇ -gal ⁇ - galactosidase
  • Female C57/BL6 p16-3MR mice are treated with doxorubicin.
  • a portion of the doxorubicin treated animals receive compound selected from the Table 1 ab and Table 1c or PBS (DOXO) daily from day 10 to day 24 post-doxorubicin treatment.
  • mice are sacrificed and fat biopsies immediately fixed and stained with a solution containing X- Gal. Untreated animals are used as negative control (CTRL).
  • CTRL negative control
  • the anti-aging properties of the compounds selected from the table 1 ab and Table 1c or compositions, comprising such compound are confirmed with the following tests: [00311] In one example, The compound selected from the table 1 ab and Table 1c is administered to groups of mice (dosages for respective groups: 0.1 mg/kg – for group #1, 1 mg/kg – for group #2, 10mg/kg – for group #3, 100mg/kg – for group #4 and 1g/kg – for group #5) intraperitoneally from weeks 12-13 and weeks 14-15. [00312] In another example, the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a dosage 20 times less than its LD50 by intraperitoneal injection.
  • the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a Maximum Tolerated dose by intraperitoneal injection.
  • the compound selected from the table 1 ab and Table 1c is administered by intraperitoneal injection (IP) in a doses: group #1 - 1 mg/kg, group #2 - 10 mg/kg, group #3 - 100 mg/kg.
  • IP intraperitoneal injection
  • the compound selected from the Table 1 ab and Table 1c is administered in dosage, regimen and root of administration as described in Example 1.
  • Administration of the compound selected from the table 1 ab and Table 1c or composition, comprising such compound produces at least one anti-aging effect, selected from the variety known in the art, including but not limited to those described in this application. [00317] Administration of the compound selected from the table 1 ab and Table 1c or composition, comprising such compound produces at least one of those effects described in Example 1 (that can be less but still statistically meaningful). [00318] Administration of the compound selected from the table 1 ab and Table 1c or composition, comprising such compound produces at least one of those effects described in Example 1 that is at least at the range of shown in Example 1 for treatment group.
  • any of the compound selected from Tables 1 ab and Table 1c are dissolved in at least one of the following ways: 1) dissolved in DMSO to 10 mM stocks and used in the final assay buffer with minimum 1% DMSO (for 10 micro M final measurements) and with 2% DMSO (for 200 micro M final measurements), 2) dissolved in DMSO to 10 mM stocks and used in the final assay buffer with minimum 0,1% DMSO 3) dissolved in water to 200 micro M stock solution. The stock solution is then used for further dilutions with assay buffer, and 4) any other dissolution known in the field.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is optionally administered in at least one of the dosages listed above once, one time a week during one month, one time a week during two months, one time a week during 3 months, daily during 1 week, 1 month, 6 months or 12 months.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is optionally administered once a week until biological age or frailty index of the subject is decreased or the treatment is repeated when the biological age or frailty index of the subject returns to the level when the first treatment had been administered or at least one symptom of aging or aging related disease or condition is alleviated.
  • the biological age or frailty index can be measured by methods known in the art, including but not limited to those described in “Identification of a blood test-based biomarker of aging through deep learning of aging trajectories in large phenotypic datasets of mice” (Avchaciov et al., 2020).
  • Example 5 Injection Formulation [00323] A composition comprising an Exemplary Injection Formulation containing a compound selected from the table 1 ab and Table 1c or composition, comprising such compound is prepared. The vial contains 5 mg of any one of the compounds selected from the table 1 ab and Table 1c or composition, comprising such compound as a powder for injection.
  • the powder for injection is reconstituted with sterile water for injections and further diluted in 0.9% sodium chloride solution for infusion. After reconstitution, each vial contains substance for injection.
  • Inactive ingredients include sodium phosphate monobasic monohydrate, sodium phosphate dibasic dihydrate, sucrose and polysorbate 80. In other example, there are no inactive ingredients, and in yet another example inactive ingredients are chosen by the expert in the field.
  • compound’s selected from the table 1 ab and Table 1c concentration is selected from the group consisting of: 0.01 mg/mL, 0.05 mg/mL, 0.1 mg/mL, 0.5 mg/mL, 1 mg/mL, 5 mg/mL, 10 mg/mL, 50 mg/mL or 100 mg/mL diluted in sodium chloride solution for infusion.
  • compound’s selected from the table 1 ab and Table 1c concentration is selected from the group consisting of: 0.01 mg/mL, 0.05 mg/mL, 0.1 mg/mL, 0.5 mg/mL, 1 mg/mL, 5 mg/mL, 10 mg/mL, 50 mg/mL or 100 mg/mL diluted in sterile water for injections.
  • compound’s selected from the table 1 ab and Table 1c concentration is selected from the group consisting of: 0.01 mg/mL, 0.05 mg/mL, 0.1 mg/mL, 0.5 mg/mL, 1 mg/mL, 5 mg/mL, 10 mg/mL, 50 mg/mL or 100 mg/mL diluted in sterile water for injections.
  • such formulations are for IV injections.
  • such formulations are for any one of other roots of injections mentioned in this application or known in the art.
  • Example 6 Tablet Formulation 10 mg of any one of the compounds selected from the table 1 ab and Table 1c or composition, comprising such compound or its orally bioavailable form or delivery device or agent providing oral bioavailability, 50 mg microcrystalline cellulose, 10 mg potato or modified starch, 10 mg polyvinylpyrolidone, 10 mg methylcellulose, 25 mg dibasic calcium phosphate, 2 mg magnesium stearate, and 1.92 g Hydroxypropylmethylcellulose are prepared in a tablet formulation.
  • Example 7 Injection formulation II [00329] 10 mg of any one of the compounds selected from Table 1 ab and Table 1c is provided in a vial in a sterile form for reconstitution as a suspension for subcutaneous injection or reconstitution as a solution with further dilution for intravenous infusion and 100 mg of mannitol as a sterile lyophilized powder.
  • Example 8 Tablet formulation II [00330] Any one of the compounds selected from the table 1 ab and Table 1c -20 mg, Ludipress -100 mg, Kollidon CL -10 mg, Magnesium stearate -10 mg and Aerosil -5 mg are prepared in a tablet formulation.
  • Example 9 Treatment for frailty reduction
  • C57BL/6J male mice aged 60 weeks (15months) are ordered from Jackson Laboratories. Animals are allowed to acclimate to the housing environment (the animal holding room) for 3 days prior to the initiation of the study. Animals are housed individually to avoid fighting. The animals have ad libitum access to standard diet (2018, Global 18% Protein Rodent Diet from Envigo++++, San Diego, CA) and acidified water (pH 2.5-3.0) throughout the study period.
  • the bedding material is hardwood chips (Sani- Chips, Cat# 7115, Envigo++++, CA, USA) and is changed Bi-weekly.
  • Four days before treatment start blood samples are collected for CBC analysis.
  • Rapamycin is prepared as 40 mg/mL stock solution in DMSO and stored at -20C. Stock solutions are diluted to 1.2 mg/ml. Final formulation - 5% Tween-80, 5% PEG-400, 3% DMSO (from stock solution) in water.
  • any one of the compounds selected from the table 1 ab and Table 1c or composition, comprising such compound is prepared as 66.7 mg/mL stock solution in DMSO and stored at -20C. Stock solutions are diluted to 2 mg/ml. Final formulation - 5% Tween-80, 5% PEG-400, 3% DMSO (from stock solution) in water. [00335] Control animals are treated with 5% Tween-80, 5% PEG-400, 3% DMS (from stock solution) in water.
  • Test/Control Agents are administered via intraperitoneal or intravenous or subcutaneous injection daily (Mon-Sun) or by any one of the protocols (dosage, regimen, root of administration) described in Example 1 or in other parts of this application. Animals’ body weights are taken on Day-3 for randomization purposes and animals are monitored by weekly body weights throughout the entire study duration. Detailed clinical observations are performed weekly and recorded. [00337] Every two weeks blood samples are collected and analyzed on hematology analyzer. The blood (120 ⁇ L) is collected into EDTA tubes via submandibular or facial vein using a lancet. [00338] The mean value of bioage, frailty index or dFI in treatment group is less in comparison to the control group.
  • At least one aging hallmark in the treatment group is reversed or alleviated.
  • At least one parameter measured as shown in Example 1 indicates anti-aging effect or senolytic effect of compound of this application.
  • compound selected from the table 1 ab and Table 1c or composition, comprising such compound is dissolved in DMSO to 10 mM stocks and used with minimum 1% DMSO (for 10 micro M final measurements) and with 2% DMSO (for 200 micro M final measurements) or compounds are dissolved in water to 200 micro M stock solution. The stock solution is then used for further tests.
  • compound selected from the table 1 ab and Table 1c or composition, comprising such compound is dissolved in sterile water to 10 mM stocks or compounds are dissolved in water to 200 micro M stock solution. The stock solution is then used for further tests.
  • the compound selected from the table 1 ab and Table 1c is administered to groups of mice (dosages for respective groups: 0.1 mg/kg – for group #1, 1 mg/kg – for group #2, 10mg/kg – for group #3, 100mg/kg – for group #4 and 1g/kg – for group #5) intraperitoneally from weeks 12-13 and weeks 14-15.
  • the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a dosage 20 times less than its LD50 by intraperitoneal injection.
  • the compound, selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in a Maximum Tolerated dose by intraperitoneal injection.
  • the compound selected from the table 1 ab and Table 1c is administered by intraperitoneal injection (IP) in a doses: group #1 - 1 mg/kg, group #2 - 10 mg/kg, group #3 - 100 mg/kg. Table 2. Complete blood counts parameters measured in the experiments, which are used for calculation of marker of biological age.
  • dFI Dynamical frailty index
  • MA0071 males, diamonds
  • MA0071 females, circles
  • MA0072 triangles
  • the black curved dashed line is the exponential fit in the age groups younger than the average lifespan of NIH Swiss mice (indicated by the dashed vertical line).
  • Stars mark the average dFI in age-matched groups of frail animals from the MA0073 cohort. All data are presented as mean ⁇ SEM.
  • dFI is associated with the number of senescent cells [00346] Total flux (TF) in log scale representing p16-dependent luciferase reporter activity as a quantitative indicator of senescent cells is shown in Figure 15: statistically significant correlations with age (a) and with dFI (b) in old mice (> 50 weeks). dFI is associated with the remaining lifespan Table 3. dFI is associated with the remaining lifespan [00347] Spearman’s rank-order correlation values and the corresponding p-values (in parentheses) for dFI with lifespan. Analysis is shown for two cohorts: Cohort 1 includes all animals with mortality data, Cohort 2 includes the subset of animals from Cohort 1 for which IGF1 measurements were available.
  • dFI correlates with Physiological Frailty index
  • PFI physiological frailty index
  • Figure 16 shows the predicted change in dFI under compound treatment.
  • Example 10 Use for anti-aging treatment in humans
  • a composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered orally.
  • composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered IV.
  • composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered by the root and in a formulation selected from this application.
  • a composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered in the dosage of 0,1 mg.
  • a composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered in the dosage of 1 mg.
  • a composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered in the dosage of 10 mg.
  • a composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered in the dosage of 25 mg.
  • a composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered in the dosage of 50 mg. In some examples, a composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered in the dosage of 100 mg. In some examples, a composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered in the dosage of 250 mg. In some examples, a composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered in the dosage of 500 mg. In some examples, a composition comprising any one of the compounds selected from Table 1 ab and Table 1c is administered in the dosage of 1000 mg.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered by the root of administration and in the highest dosage as it was in Phase 1 clinical trial of such compound.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered by the root of administration and in the dosage selected from the group of dosages used in Phase 1 (if any) clinical trial of such compound.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered by the root of administration and in the highest dosage as it was in Phase 2 (if any) clinical trial of such compound.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered by the root of administration and in the lowest dosage as it was in Phase 2 (if any) clinical trial of such compound.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered by the root of administration and in the highest dosage as it was in Phase 2 (if any) clinical trial of such compound.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered by the root of administration and in the dosage as it was approved for other indication.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in its Maximum Tolerated Dose.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in dosage corresponding to 75% of its Maximum Tolerated Dose.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in dosage corresponding to 50% of its Maximum Tolerated Dose.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in dosage corresponding to 25% of its Maximum Tolerated Dose.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in dosage corresponding to 10% of its Maximum Tolerated Dose. In yet another example, the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in dosage corresponding to 5% of its Maximum Tolerated Dose. In yet another example, the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in dosage corresponding to 1% of its Maximum Tolerated Dose. In yet another example, the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in dosage corresponding to 0,1 % of its Maximum Tolerated Dose.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in dosage corresponding to 0,01 % of its Maximum Tolerated Dose.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered in the regimen as it was administered at the highest phase of its clinical trials known in the art.
  • the dosage is calculated to get the dosage selected from the group consisting of: 0.5 mg, 0.2 mg, 0.05 mg, 0.005 mg, 0.001 mg, 0.1 mg, 1mg, 5 mg or 10 mg of any one of the compounds selected from the Table 1 ab and Table 1c per 1 kg of body weight.
  • the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is optionally administered orally, intravenously, intra-arterially or intra-coronary arterially. [00364] In some examples, the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered once [00365] In some examples, the compound selected from the table 1 ab and Table 1c or composition, comprising such compound is optionally administered in at least one of the dosages listed in this application in the regimen selected from the group consisting of: once, one time a week during one month, one time a week during two months, one time a week during 3 month, daily during 1 week, daily during 1 month, daily during 6 months or daily during 12 months, every 12 hours lifelong, 1 week daily having 1 week off the drug lifelong, 1 week daily having 1 month off the drug lifelong, 1 month daily having 1 year off the drug lifelong.
  • the treatment is optionally repeated when the biological age of the subject returns to the level when the first treatment was administered. [00367] In some examples, the treatment is optionally repeated when the symptom of aging or aging related disease or condition has returned to the level existed just before the first treatment. [00368] In yet another example, composition comprising any one of the compounds selected from the table 1 ab and Table 1c or composition, comprising such compound is administered orally every 24 hours for 1 week, 1 month and 3 months and is repeated when the biological age of the subject returns to the level when the first treatment had been administered.
  • composition comprising any one of compound selected from the table 1 ab and Table 1c or composition, comprising such compound is administered orally every 12 hours for 1 week, 1 month and 6 months and is repeated when the biological age of the subject returns to the level when the first treatment had been administered.
  • composition comprising any one of the compounds selected from the table 1 ab and Table 1c or composition, comprising such compound is administered by the protocol selected from this application.
  • the compounds are administered to subject of 75 kg weight in a tablet formulation, where the tablet comprises: any one of the compounds selected from Table 1 ab and Table 1c –in the dosage selected from the group comprising: 1 mg, 10 mg, 50 mg, 100 mg, 200 mg, 500 mg, 750 mg, 1 g and 1 Ludipress -17 mg, Kollidon CL -2 mg, Magnesium stearate -2 mg, Aerosil -1 mg.
  • the patient is healthy, aged or has at least one aging related disease, disorder or decline. For example, for the component of frailty “hands grip”, the hands grip force is measured by the means known in the art.
  • RT reaction time
  • the score on this task is the mean response time in milliseconds across trials which contained matching pairs. See for details e.g. Cognitive Test Scores in UK Biobank: Data Reduction in 480,416 Participants and Longitudinal Stability in 20,346 Participants (Donald M. Lyall et al., 2016). After the treatment, the force of hands grip is increased and the time of mean response time in test of symbol matching is decreased.
  • any one of the compounds selected from Table 1 ab and Table 1c is used to prevent and treat frailty.
  • the frailty index of a subject can be measured by many ways known in the art, including but not limited to as described in Rockwood et al., 2005, Searle et al.2008 and Rockwood et al., 2017.
  • Other anti-aging effects are measured in many ways known in the art including but not limited to those described in Justice et al., 2016, an effect against particular diseases – by the methods, tests and equipment known to the expert in the particular disease etc.
  • Anti-aging effects are also identified as a change of at least one of the biomarker of aging or aging related decline into more juvenile state or the delay in progression into more biological elder state in comparison with untreated.
  • Anti-aging effects are also identified as an alleviation of at least one symptoms of aging or aging related disease or condition.
  • mice are treated with vehicle and any one of the compounds selected from the table 1 ab and Table 1c or composition, comprising such compound, PO administration, every 6h at 0.05 mg/kg, 0.5 mg/kg, 1 mg/kg or 10 mg/kg.
  • the mice are treated by the protocol (dosage, regimen and root of the administration) described in Example 1.
  • any one of the compound selected from the table 1 ab and Table 1c or composition, comprising such compound or any other compound of this disclosure is injected, optionally 0.001 mg/kg, 0.025 mg/kg, 0.25 mg/kg, 0.01 mg/kg, 0.5 mg/kg, 1 mg/kg, 2.5 mg/kg, 5 mg/kg, 10 mg/kg, 25 mg/kg or 50 mg/kg, 2 times a day by IV.
  • subcutaneous or intraperitoneal injection can be done.
  • injection of any one of the compound selected from the table 1 ab and Table 1c or composition, comprising such compound or any other compound of this disclosure in the same dosage as shown above in this example is administered subcutaneously or orally.
  • the animals are treated once, or daily: for 1 week, for 2 weeks, for 1 month, for 3 months or for 6 months.
  • weight and fasting blood glucose, insulin, triglycerides, and leptin levels are assessed and glucose/insulin tolerance tests are performed.
  • animals treated with the agent or combination of this application will have healthier levels of the parameter selected from the group consisting of: weight, fasting blood glucose, insulin, triglycerides, leptin levels and glucose/insulin tolerance.
  • EXAMPLE 12 Animal experiment on aging related diseases and disorders [00383] This experiment is conducted on a relevant species (for example, mice, rat etc.) and disease model of a particular aging related disease, disorder or decline. [00384] As non-limiting examples, it could be animal models known in the art, e.g.
  • models of disease or condition selected from the group consisting of: Alzheimer's disease, Parkinson's disease, cataracts, macular degeneration, glaucoma, atherosclerosis, acute coronary syndrome, myocardial infarction, stroke, recovery after stroke, PSD-95 decrease, hypertension, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), type 1 diabetes, obesity, fat dysfunction, coronary artery disease, cerebrovascular disease, periodontal disease, cancer treatment-related disability, chemotherapy treatment-related disability, chemotherapy treatment-related frailty, frailty, radiotherapy and other radiation related disability, chemotherapy treatment-related frailty, cancer treatment-related atrophy, cancer treatment-related fibrosis, brain injury, heart injury, and therapy-related myelodysplastic syndrome, accelerated aging, accelerated aging disease, Hutchinson-Gilford progeria syndrome, Werner syndrome, Cockayne syndrome, exroderma pigmentosum, ataxia telangiectasia, Fanconi
  • the animal models for senescence associated diseases and conditions are described in patent application EP3099380A1 “Methods and compositions for killing senescent cells and for treating senescence-associated diseases and disorders”, optionally the dosage, regimen and root of administration is selected from the Example 1.
  • the dosage and regimen are taken as defined as expert in the field.
  • the protocol of compound selected from the table 1 ab and Table 1c is taken from the Example 1.
  • the monitoring of efficacy of the compound selected from the table 1 ab and Table 1c tested in such model can be trivially done using the methods known for the respective animal model.
  • EXAMPLE 12.2 Stroke recovery Humans [00389] Patients who had ischaemic stroke and hemiplegia or hemiparesis, have Fugl-Meyer motor scale (FMMS) scores of 55 or less or any other post-stroke syndromes known to the experts in the field are studied in this example. [00390] In one example, the compound selected from the table 1 ab and Table 1c is used by in the formulation, root of administration, dosage and regimen selected from the Example 10. [00391] The primary outcome measure is the change on the FMMS between day 0 and day 90 after the start of the study drug in comparison with placebo. Participants, carers, and physicians assessing the outcome are masked to group assignment.
  • FMMS Fugl-Meyer motor scale
  • any one of the compounds selected from the table 1 ab and Table 1c or composition, comprising such compound or placebo for 3 months are administered starting 5–10 days after the onset of stroke by the route, regimen and dosage selected from the described in any one of the Examples 1, 4,9, 10, 11, 12.1 or 12.2 or by the route, regimen and dosage as it is used for its primary indication or by the route, regimen as it is used for its primary indication, but the dosage is about selected from the group consisting of: 0,1; 0,5; 2 times more, 5 times more, 10 times, 50 times more in comparison with the effective dosage of its primary indication.
  • Footprints are captured by a high- speed camera and analyzed on the computer using the Catwalk XT 8.1 software.
  • the animals are trained for 3 days before surgery and tested at days 1, 7, 14, 28, 42 after stroke.
  • the max contact area and the print length are used to evaluate the influence of the Compound selected from the table 1 ab and Table 1c treatment to the mice after stroke.
  • Compound selected from the table 1 ab and Table 1c Increases Neuronal Regeneration and Improved Functional Recovery After Stroke
  • Effect of the treatment on biological age is estimated based on changes in standard blood count (Antoch et al, 2017), DNA methylation (Stubbs et al., 2017; Horvath, 2013), lifespan (Harrison et al., 2009), and healthspan assessed by frailty index.
  • a frailty index is created by counting the accumulation of deficits in health across many systems in the body. Deficits measured to construct a frailty index include a large number of health-related variables related to the function of systems that are known to change with age in both human and animal models (Parks et al., 2012).
  • variables provide information about the following: (a) activity, including distance moved, velocity of movement and rearing frequency; (b) hemodynamic status, including heart rate, systolic and diastolic blood pressure; (c) body composition, including body mineral content, percent body fat and percent lean tissue; and (d) basic metabolism and organ function, including serum electrolyte levels, hematocrit and urea levels clinical signs, symptoms, diseases, and laboratory and radiographic abnormalities.
  • Cognitive score is assessed using Barnes maze, activity chamber and fear-conditioning tests.
  • the Barnes maze test consists of a large circular maze containing 40 holes and elevated approximately 40 cm above the floor.
  • the escape hole consists of a long PVC elbow joint connector that was similar in texture and color to the maze.
  • the escape hole position is fixed within a day but changed for each successive day of testing.
  • the starting location for the mouse is rotated relative to the escape hole position.
  • the maze and escape hole are thoroughly cleaned to remove any cues that might affect performance in subsequent trials.
  • mice are given 90 s for each trial to identify the escape hole by jumping in or identifying the hole with extended/overt head pokes.
  • An activity chamber Med Associates Inc., St. Albans, VT is used to evaluate general locomotor activity and exploratory behavior in a novel environment.
  • each mouse is placed in the foot-shock chamber with a front-mounted camera to visualize various pain sensitivity parameters, which encompasses four clearly defined behaviors, ‘flinch’, ‘run’, ‘vocalization’, and ‘two- paw jump’.
  • One-second shocks of various intensities are delivered each spaced by 30 s of recovery time.
  • Thresholds reflected the minimum shock intensity at which each behavior was observed using the front- mounted camera video.
  • Example 14 COVID-19 experiments In vitro experiments [00406] To investigate anti-SARS-CoV-2 activity of any one of the compounds selected from the table 1 ab and Table 1c or composition, comprising such compound in vitro, Vero E6 cells (ATCC-1586) are used. The cells are grown in Dulbecco's Modified Eagle Medium (DMEM) (Sigma Aldrich, Boston, MA, USA) supplemented with 5% fetal bovine serum (Logan, UT, USA) at 37°C and 5% CO2. [00407] Cytotoxicity of the compounds in Vero cells is determined by the CCK8 assay.
  • DMEM Dulbecco's Modified Eagle Medium
  • Vero cells are seeded into 96-well plates at a density of 1 ⁇ 104 cells/well and grown for 24 hours. Cells are then infected with SARS-CoV-2 at a multiplicity of infection (MOI) of 0.01 (100 PFU/well) for 2 hours at a temperature of 37°C. Virus input is washed with DMEM and the cells are then treated with medium containing test compounds at various concentrations (3-fold dilutions at seven different concentrations starting from ⁇ 50/10) for 24 or 48 hours.
  • MOI multiplicity of infection
  • Vero cells are grown as described above and are pretreated with various concentrations of the compounds ((3-fold dilutions at seven different concentrations starting from ⁇ 50/10) for 2 hours, after that drug containing medium is removed and SARS-CoV-2 virus-containing medium is added (as described for the treatment study) for 2 hours. Following this, the virus-containing medium is removed and replaced with fresh medium that does not contain drugs or viruses.
  • Efficacies are evaluated by quantification of viral copy numbers in the cell supernatant via quantitative real-time RT-PCR (qRT-PCR) (as described in [Huang C, Wang Y, Li X, et al.
  • All infections are performed in an animal biosafety level 3 facility using appropriate practices, including a HEPA-filtered bCON caging system, HEPA-filtered powered air- purifying respirators (PAPRs), and Tyvek suiting. All animals are grown to 10 weeks of age prior to use in experiments. The animals are anesthetized using a mixture of xylazine (0.38 mg/mouse) and ketamine (1.3 mg/mouse) in a 50- ⁇ l total volume by intraperitoneal injection. The mice are inoculated intranasally with 50 ⁇ l of either PBS or 1 ⁇ 105 PFU of rMA15 SARS-CoV, after which all animals are monitored daily for weight loss.
  • Test compounds are administered - every day via i.p route in the dosage which is used for the successful efficacy tests made at the same or alike animal for such drug’s initial indication -or by the route, regimen and dosage which is used for the efficacy tests made at the same animal.
  • the protocol of compound administration is selected from the Example 1.
  • - Mice are euthanized at days 2, 5, or 9 post infection, and lung tissue was harvested for further analysis.
  • Fifty percent tissue culture infectious dose (TCID50) values from SARS-CoV(MA15)- infected lungs is calculated by infecting multiple replicates of Vero E6 cells plated on 96-well plates.
  • Serial dilutions (1:5) are performed for the virus-containing lung lysates in Vero E6 culture medium (complete MEM) such that at least the last two dilutions had no detectable virus in any of the replicates.
  • the infection proceeds for 3 days before cells are fixed with 4% PFA and stained with 0.05% crystal violet in 20% methanol.
  • Example 15 Cancer supportive care treatment 15.1
  • Animals are treated with a dose corresponding to LD10 or to LD25 of a chemotherapy agent, for example but not limited to one selected from the following group consisting of: aminoglutethimide, amsacrine, anastrozole, asparaginase, bcg, bicalutamide, bleomycin, bortezomib, buserelin, busulfan, campothecin, capecitabine, carboplatin, carfilzomib, carmustine, chlorambucil, chloroquine, cisplatin, cladribine, clodronate, colchicine, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, demethoxyviridin, dichloroacetate, dienestrol, diethylstilbestrol, do
  • a chemotherapy agent for example but not limited to
  • Frailty Index is measured as described in Animals are left for one month of recovery. FI measurement is repeated. Animals are stratified in two groups: the control and the treatment groups. Treatment by a compound selected from Table 1 ab and Table 1c is performed for 4-8 weeks followed by FI measurement, followed by another dose of LD10 or LD25 of chemotherapy agent. [00415] In some examples, compound selected from Table 1 ab and Table 1c is administered by the root of administration, dosage, formulation selected from the example 1. [00416] Survival is evaluated. Animals in treatment group demonstrate better survival. Clinical [00417] Patients undergoing chemotherapy are enrolled. After the first round of chemotherapy FI is evaluated. Patients are stratified in placebo and treatment groups.
  • Treatment groups receive a compound selected from the Table 1 ab and Table 1c or from any other compounds described herein. Patients receive the second round of chemotherapy treatment. Rate of withdrawal and dose reduction is evaluated. Patients in the treatment group have lower rates of withdrawal and dose reduction. [00418] In some examples, Any one of the compounds selected from Table 1 ab and Table 1c or any other compound described herein is administered by PO administration, every 24h in a dosage of 0.001 mg/kg, 0.005 mg/kg, 0.01 mg/kg, 0.05 mg/kg, 0.5 mg/kg, 1 mg/kg or 10 mg/kg. [00419] In some examples, Any one of the compounds selected from Table 1 ab and Table 1c or any other compound described herein may be administered with food.
  • the compound could be mixed with feed at a dosage of 0.01 mg/kg, 0.1 mg/kg, 1 mg/kg, 5 mg/kg, 10mg/kg, 20mg/kg or 50mg/kg.
  • Any one of the compounds selected from Table 1 ab and Table 1c or any other compound described herein may be administered by IV injection, for example 0.025 mg/kg, 0.25 mg/kg, 0.01 mg/kg, 0.5 mg/kg, 1 mg/kg, 2.5 mg/kg, 5 mg/kg, 10 mg/kg, 25 mg/kg or 50 mg/kg -1 time a day.
  • any one of the compounds selected from Table 1 ab and Table 1c or any other compound described herein may be administered subcutaneously or orally in the same dosage as shown above in this example.
  • the protocol of administration is selected from the Example 10.
  • Patients in the treatment group have lower rates of withdrawal and dose reduction. 15.2 Use together with immunotherapy
  • any one of the compounds selected from Table 1 ab and Table 1c , or composition, comprising such compound, other compound or composition disclosed herein can be used or administered prior the immunotherapy in the formulation, dosage and regimen as shown in any one of the Examples 10 above or any other part of this disclosure or as defined by practitioner.
  • any one of the compounds selected from Table 1 ab and Table 1c or composition, comprising such compound can be used or administered concomitantly with the immunotherapy in the formulation, dosage and regimen as shown in Examples 10.
  • any one of the compounds selected from Table 1 ab and Table 1c or composition, comprising such compound can be used or administered as defined by practitioner.
  • the compound of this disclosure is administered prior the treatment by the immunotherapy in regimen and dosage effective for changing at least one of aging biomarkers of the patient in younger or more healthy state, and immunotherapy is used soon after such biomarker was improved.
  • the biomarker or aging related biomarker known for practitioners as being predictive for the efficacy of the particular immunotherapy is measured.
  • Example 16 Direct anti-cancer action Primary Adenocarcinoma NSCLC Xenograft Model [00424] Tumor tissue is excised from a patient with a poorly differentiated adenocarcinoma. This tumor tissue is implanted subcutaneously in the flank of a SCID mouse and passaged twice before compound testing.
  • a compound selected from Table 1 ab and Table 1c is dosed as a solution alone in the dosage, regimen and route of administration for compounds is as in Example 1 or on 4 consecutive days per weekly cycle (qd4, dosed on days 1, 2, 3 and 4 each week).
  • a control group receives vehicle alone (10% Vitamin E TPGS in water, po qd4). All drug treatment is stopped on Day 28. Vehicle, and compound treatment groups are sacrificed and the remainder monitored for a further 40 days to assess tumor re-growth. Tumor size in treatment group are less and other cancer symptoms are alleviated.
  • the transgenic mouse comprises a p16 Ink4a promoter operatively linked to a trimodal fusion protein for detecting senescent cells and for selective clearance of senescent cells in these transgenic mice.
  • the promoter, p16 Ink4a which is transcriptionally active in senescent cells but not in non-senescent cells (see, e.g., Wang et al, J. Biol. Chem. 276:48655-61 (2001); Baker et al, Nature 479:232-36 (2011)), is engineered into a nucleic acid construct.
  • 3MR tri-modality reporter
  • LOC Renilla luciferase
  • mRFP monomeric red fluorescence protein
  • tTK truncated herpes simplex virus
  • GCV ganciclovir
  • Insertion of the 3MR cDNA also result in the occurrence of a stop codon in the pl9ARF reading frame in exon 2, thereby preventing full-length pi 9ARF expression from the BAC as well.
  • the p16Ink4a gene promoter (approximately 100 kilobase pairs) is introduced upstream of a nucleotide sequence encoding a trimodal reporter fusion protein.
  • a truncated pi 6Ink4a promoter may be used (see, e.g., Baker et al., Nature, supra; International Application Publication No.WO2012/177927; Wang et al, supra).
  • the expression of 3MR is driven by the p16Ink4a promoter in senescent cells only.
  • the detectable markers, LUC and mRFP permitted detection of senescent cells by bio luminescence and fluorescence, respectively.
  • the expression of tTK permitted selective killing of senescent cells by exposure to the pro-drug ganciclovir (GCV), which is converted to a cytotoxic moiety by tTK.
  • GCV pro-drug ganciclovir
  • mice are randomized into doxorubicin + the compound selected from the table 1 ab and Table 1c treated or doxorubicin only treated groups. Senescence is induced by intraperitoneal administration of doxorubicin at 10 mg/kg to the mice ten days prior to administration of the compound selected from the table 1 ab and Table 1c (Day -10).
  • R A is analyzed for mR A levels of endogenous senescence markers (e.g., p21 , p16 INK4a (pi 6), and p53) and SASP factors (e.g., mmp-3 and IL-6) relative to actin mRNA (control for cDNA quantity) using the Roche Universal Probe Library for real-time PCR assay.
  • endogenous senescence markers e.g., p21 , p16 INK4a (pi 6), and p53
  • SASP factors e.g., mmp-3 and IL-6
  • actin mRNA control for cDNA quantity
  • ALEXA FLUOR® dye-labeled secondary goat anti-rabbit antibody (Life Technologies) and counterstained with 4',6-diamidino-2-phenylindole (DAPI) (Life Technologies).
  • DAPI 4',6-diamidino-2-phenylindole
  • the number of positive cells is calculated using Image J image processing program (National Institutes of Health, see Internet at imagej.nih.gov/ij/index.html) and represented as a percentage of the total number of cells.
  • Upon collection, fat biopsies are immediately fixed in 4% formalin and then stained with a solution containing X-gal to detect the presence of senescence- associated ⁇ -galactosidase ( ⁇ -gal).
  • Fat biopsies are incubated overnight at 37 °C in X- gal solution and are photographed the next day. Fat biopsies from untreated animals are used as a negative control (CTRL).
  • CRL negative control
  • EXAMPLE 18 ANIMAL MODEL OF OSTEOARTHRITIS The two treatment studies are designed to determine the effect of removing senescent cells in an animal model of osteoarthritis. Parallel studies can be performed. One study investigates the effect of eliminating senescent cells with the compound selected from the table 1 ab and Table 1c in 3MR mice. Mice should go under surgery to cut the anterior cruciate ligament of one rear limb to induce osteoarthritis in the joint of that limb.
  • mice groups receive 0.01 ⁇ g, 0.1 ⁇ g, 1 ⁇ g, 2.5 ⁇ g, 5 ⁇ g, 10 ⁇ g and 100 ⁇ g of the compound selected from the table 1 ab and Table 1c to the operated knee by intra- articular injection, qd for 5 days, with a 2 nd treatment (the same dose of the compound selected from the table 1 ab and Table 1c qd for 5 days) during week 4 post-surgery.
  • operated joints of the mice are monitored for presence of senescent cells, assessed for function, monitored for markers of inflammation, and underwent histological assessment.
  • C57BL/6J mice should go under surgery to cut the anterior cruciate ligament of one rear limb to induce osteoarthritis in the joint of that limb.
  • seven 3MR mice groups receive each group – particular dosage: 0.01 ⁇ g, 0.1 ⁇ g, 1 ⁇ g, 2.5 ⁇ g, 5 ⁇ g, 10 ⁇ g and 100 ⁇ g of the compound selected from the table 1 ab and Table 1c per operated knee by intra-articular injection, qod for 2 weeks.
  • the protocol of administartion of compound selected from the table 1 ab and Table 1c is selected from the Example 1.
  • compound selected from the table 1 ab and Table 1c is administered During week 3 and week 4 post- surgery, the rest of the protocol of administartion of compound selected from the table 1 ab and Table 1c is selected from the Example 1. At the end of 4 weeks post-surgery, joints of the mice are monitored for presence of senescent cells, assessed for function, monitored for markers of inflammation, and underwent histological assessment.
  • RNA from the operated joints of mice from the compound selected from the table 1 ab and Table 1c treated mice are analyzed for expression of SASP factors (mmp3, IL-6) and senescence markers (pi 6).
  • qRT-PCR is performed to detect mRNA levels.
  • the treatment with the compound selected from the table 1 ab and Table 1c clears senescent cells from the joint.
  • RNA from the operated joints of mice are also analyzed for expression of type 2 collagen and compared with expression of actin as a control.
  • the treatment with the compound selected from the table 1 ab and Table 1c in mice that have undergone osteoarthritis surgery drives collagen production as compared to untreated mice. Function of the limbs are assessed 4 weeks post-surgery by a weight bearing test to determine which leg the mice favored.
  • the mice are allowed to acclimate to the chamber on at least 3 occasions prior to taking measurements. Mice are maneuvered inside the chamber to stand with 1 hind paw on each scale.
  • the weight that is placed on each hind limb is measured over a 3- second period. At least 3 separate measurements are made for each animal at each time point. The results are expressed as the percentage of the weight placed on the operated limb versus the contralateral unoperated limb.
  • the untreated mice that have undergone osteoarthritis surgery favor the unoperated hind limb over the operated hind limb ( ⁇ ).
  • clearing senescent cells with the compound selected from the table 1 ab and Table 1c abrogates this effect in mice that have undergone surgery (V).
  • the compound selected from the table 1 ab and Table 1c abrogates this effect in mice that have undergone surgery (V).
  • mice The function of the limbs is also assessed at 4 weeks post-surgery by hotplate analysis to show sensitivity and reaction to pain stimulus.
  • a mouse is placed on a hotplate at 55°C. When placed on the hot surface of the plate, mice will lift their paws and lick them (paw-lick response) due to attainment of pain threshold. The latency period for the hind limb response (paw-lick response) is recorded as response time.
  • the untreated mice that have undergone osteoarthritis surgery have an increased response time as compared to normal mice that have not been surgically altered. However, treatment of mice that have undergone osteoarthritis surgery with the compound selected from the table 1 ab and Table 1c decreases the response time in a significant manner.
  • Example 20 EFFECT COMPOUND SELECTED FROM TABLE 1 ab and Table 1c IN PULMONARY DISEASE MODELS
  • bleomycin injury model for idiopathic pulmonary fibrosis, mice develop lung fibrosis within 7-14 days after bleomycin treatment.
  • Bleomycin is administered to anesthetized 6-8 week old 3MR mice by intratracheal aspiration (2.5U/kg of bleomycin in 50 ⁇ PBS) using a microsprayer syringe (Penn- Century, Inc.) as described in Daniels et al. (2004, J. Clin.
  • mice are administered saline.
  • Compound selected from the table 1 ab and Table 1c in different dosages (0.01 mg/kg – for group #1, 0.1 mg/kg – for group #2, 1 mg/kg – for group #3, 10 mg/kg – for group #4, 25mg/kg – for group #5, 50 mg/kg – for group #6, 100 mg/kg – for group #7, 500 mg/kg – for group #8 in PBS) is administered.
  • Compound selected from the table 1 ab and Table 1c is administered in dosage and root of administration selected from the Example 1.
  • the day following bleomycin treatment Compound selected from the table 1 ab and Table 1c is administered via intraperitoneal injection in the dosage 10-fold less than LD50. In yet another example, the day following bleomycin treatment, Compound selected from the table 1 ab and Table 1c is administered via intraperitoneal injection in the dosage 100-fold less than LD50. In yet another example, the day following bleomycin treatment, Compound selected from the table 1 ab and Table 1c is administered in dosage, regimen and root of administration selected from the Example 1. In one example, 3MR mice is treated via intraperitoneal injection with Compound selected from the table 1 ab and Table 1c for 5 consecutive days, followed by 5 days of rest, followed by a second treatment cycle of 5 consecutive days.
  • 3MR mice is treated with Compound selected from the table 1 ab and Table 1c in dosage, regimen and root of administration selected from the Example 1.
  • Untreated mice receive an equal volume of vehicle.
  • lung function is assessed by monitoring oxygen saturation using the MouseSTAT PhysioSuite pulse oximeter (Kent Scientific). Animals are anesthetized with isoflurane (1.5%) and a toe clip is applied. Mice are monitored for 30 seconds and the average peripheral capillary oxygen saturation (Sp0 2 ) measurement over this duration is calculated.
  • the bleomycin administration significantly reduce Sp02 levels in vehicle treated mice, and removal of senescent cells results in higher Sp02 levels, which approach normal levels at 21 days post bleomycin administration.
  • AHR airway hyper-reactivity
  • mice At 21 days post-bleomycin treatment, airway hyper-reactivity (AHR) of mice is examined.
  • AHR of mice is measured by methacholine challenge while other parameters of lung function (airway mechanics, lung volume and lung compliance) is determined using a SCIREQ flexiVent ventilator.
  • airway resistance elastance
  • compliance of mice are assessed at baseline and in response to increasing concentrations of methacholine (0 to 50 mg/ml in PBS) delivered via nebulization (AeroNeb) as described in Aravamudan et al. (Am. J.
  • SCIREQ FlexiVentTM ventilator and lung mechanics system
  • bleomycin administration increase lung elastance
  • Compound selected from the table 1 ab and Table 1c treatment reduces lung elastance.
  • the bleomycin administration reduces static compliance and (dynamic) compliance in vehicle treated mice. Clearance of senescent cells with Compound selected from the table 1 ab and Table 1c in bleomycin exposed mice improves compliance values.
  • mice are euthanized by i.p injection of pentobarbital. Bronchoalveolar lavage (BAL) fluids and lungs is obtained and analyzed. Hydroxyproline content of lungs is measured as described in Christensen et al. (1999, Am . J. Pathol.155: 1773-1779), and quantitative histopathology is performed. RNA is extracted from lung tissue to measure senescence cell markers by qRT-PCR in treated and control mice. The effect of clearance of senescence cells in the bleomycin induced lung injury model of IPF may also be studied in INK- ATT AC transgenic mice in the study design described above.
  • BAL Bronchoalveolar lavage
  • INK- ATT AC p16 Ink4a apoptosis through targeted activation of caspase
  • transgenic mice have an FK506-binding protein (FKBP)-caspase 8 (Casp8) fusion polypeptide under the control of the p16 Ink4a promoter (see, e.g., Baker et al, Nature, supra; Int'l Patent Application Publication No. WO/2012/177927).
  • FKBP FK506-binding protein
  • Casp8 caspase 8
  • a synthetic drug that induces dimerization of a membrane bound myristoylated FKBP-Casp8 fusion protein senescent cells specifically expressing the FKBP-Casp8 fusion protein via the p16 Ink4a promoter undergo programmed cell death (apoptosis) (see, e.g., Baker, Nature, supra, Figure 1 therein).
  • a second study also assesses the effect of clearance of senescence cells using a compound selected from the table 1 ab and Table 1c in C57BL6/J mice that have bleomycin induced lung injury. Bleomycin is administered to 6 week old C57BL6/J mice as described above.
  • a compound selected from the table 1 ab and Table 1c is administered during the first and third week post-bleomycin treatment in the dosage and root of administration selected from the Example 1.
  • Control mice are treated with vehicle.
  • clearance of senescent cells and lung function/histopathology is assessed.
  • a second animal model for pulmonary diseases e.g., COPD
  • mice are exposed to cigarette smoke.
  • the effect of a Compound selected from the table 1 ab and Table 1c on the mice exposed to smoke is assessed by senescent cell clearance, lung function, and histopathology.
  • the COPD protocol is adapted from the COPD core facility at Johns Hopkins University (at Internet site web.jhu.edu/Biswal/exposure_core/copd.html#Cigarette_Smoke) (Rangasamy et al, 2004, J. Clin. Invest. 114: 1248-1259; Yao et al, 2012, J. Clin.
  • mice receive a total of 6 hours of cigarette smoke exposure per day, 5 days a week for 6 months.
  • Each lighted cigarette (3R4F research cigarettes containing 10.9 mg of total particulate matter (TPM), 9.4 mg of tar, and 0.726 mg of nicotine, and 11.9 mg carbon monoxide per cigarette [University of Kentucky, Lexington, KY]) is puffed for 2 seconds and once every minute for a total of 8 puffs, with the flow rate of 1.05 L/min, to provide a standard puff of 35 cm 3 .
  • the smoke machine is adjusted to produce a mixture of side stream smoke (89%) and mainstream smoke (11%) by smoldering 2 cigarettes at one time.
  • the smoke chamber atmosphere is monitored for total suspended particulates (80-120 mg/m 3 ) and carbon monoxide (350 ppm).
  • INK-ATTAC and (10) 3 MR mice are treated with compound selected from the table 1 ab and Table 1c (3x per week) (5 consecutive days of treatment followed by 16 days off drug, repeated until the end of the experiment), respectively.
  • INK-ATTAC and (10) 3 MR mice are treated with compound selected from the table 1 ab and Table 1c administered in the dosage and root of administration selected from the Example 1 (5 consecutive days of treatment followed by 16 days off drug, repeated until the end of the experiment), respectively.
  • mice The remaining 30 mice (15 INK-ATTAC and 153 MR) were evenly split with 5 of each genetically modified strain placed into treatment groups.
  • the dosage and root of administration is selected from the Example 1.
  • An additional 70 animals that did not receive exposure to cigarette smoke are used as controls for the experiment. After two months of cigarette smoke exposure, lung function is assessed by monitoring oxygen saturation using the MouseSTAT PhysioSuite pulse oximeter (Kent Scientific). Animals are anesthetized with isoflurane (1.5%) and the toe clip was applied.
  • mice are monitored for 30 seconds and the average peripheral capillary oxygen saturation (Sp02) measurement over this duration is calculated.
  • the clearance of senescent cells via Compound selected from the table 1 ab and Table 1c results in statistically significant increase in Sp0 2 levels in mice after 2 months of cigarette smoke exposure compared to untreated controls.
  • AHR airway hyper-reactivity
  • mice are killed by i.p. injection of pentobarbital for in-depth analysis of lung histopathology as previously described (Rangasamy et al, 2004, J. Clin. Invest.114: 1248-1259).
  • lungs are inflated with 0.5% low-melting agarose at a constant pressure of 25 cm.
  • Part of the lung tissue is collected for RNA extraction and qRT-PCR analysis of senescent markers.
  • Other parts of lungs are fixed in 10% buffered formalin and embedded in paraffin. Sections (5 um) are stained with hematoxylin and eosin.
  • Mean alveolar diameter, alveolar length, and mean linear intercepts are determined by computer- assisted morphometry with Image Pro Plus software (Media Cybernetics).
  • the potential therapeutic effect of clearance of senescent cells after COPD is fully developed may be assessed in 3MR or INK-ATTAC mice. Six week-old 3MR or INK-ATTAC mice are chronically exposed to cigarette smoke for 6 months as described above.
  • 3MR or INK- ATTAC mice are treated with Compound selected from the table 1 ab and Table 1c (5 consecutive days of treatment followed by 16 days off drug) in a dosage and root of administration selected from the table 1 ab and Table 1c , respectively, until 9 months following the start of smoke exposure, when assessment of senescent cell clearance, lung function, and histopathology is performed.
  • the lung function of the tretament group is improced in comparison with the control group even without the direct senolytic action of compound selected from the table 1 ab and Table 1c .
  • EXAMPLE 21 EFFECT OF THE COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c TO REDUCE CHEMOTHERAPY RELATED SIDE EFFECTS
  • paclitaxel also induces cellular senescence when administered to animals.
  • Paclitaxel induces senescence and SASP in p16-3MR transgenic mice.
  • Senescence is observed as shown by luminescence in mice treated with paclitaxel (known in the art).
  • the level of mRNA in skin is determined for each of the target genes: pi 6, 3MR transgene, and IL-6.
  • the levels of mRNA for each of pi 6, 3MR, and IL-6 increased in paclitaxel treated animals compared with vehicle treated animals.
  • paclitaxel is administered to groups of p16-3mr mice three times, every two days.
  • COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c is administered daily for three days (days 1, 2, and 3) at different dosages (0.01 mg/kg – for group #1, 0.1 mg/kg – for group #2, 1 mg/kg – for group #3, 10 mg/kg – for group #4, 25mg/kg – for group #5, 50 mg/kg – for group #6, 100 mg/kg – for group #7, 500 mg/kg – for group #8)
  • COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c is administered intraperitoneally daily for seven days after paclitaxel administration at different dosages (0.01 mg/kg – for group #1, 0.1 mg/kg – for group #2, 1 mg/kg – for group #3, 10 mg/kg – for group #4, 25mg/kg – for group #5, 50 mg/kg – for group #6, 100 mg/kg – for group #7, 500
  • COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c is administered daily for seven days after paclitaxel administration at dosages and by the root of administration selected from the Example 1. Two days after the last dose of COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c , all groups of animals are housed in metabolic cages (promethion, sable systems international, Las Vegas, NV) to monitor voluntary exercise as determined by wheel counts. Data is collected and analyzed two days later. Administration of COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c restores wheel count reduction caused by chemotherapy treatment.
  • Animals are then treated with COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c via intraperitoneal injection (3 rounds administered daily for five consecutive days at the dosage 100-fold less than LD50) or vehicle.
  • Animals are then treated with COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c (3 rounds administered daily for five consecutive days at the dosage and root of administration selected from the Example 1) or vehicle.
  • Animals are then treated with COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c according to the protocol selected from the Example 1.
  • a glucose bolus is given at time zero, and blood glucose is monitored at 20, 30, 60, and 120 minutes after delivering glucose to determine glucose disposal).
  • AUC area under the curve
  • COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c -treated mice show a greater decrease in blood glucose at 0, 14, 30, 60, and 120 minutes after the administration of glucose bolus at time zero, suggesting that senescent cell clearance improves insulin sensitivity. Changes in weight, body composition, and food intake are also monitored. In this example, treatment by COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c will not alter body weight, body composition monitored by measuring percent of fat, or food intake (measured in grams per week).
  • EXAMPLE 23 COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c Sustains Cardiac Stress Resistance with Aging
  • the cohorts of INK-ATTAC transgenic mice on FVB ⁇ 129Sv/E ⁇ C57BL/6 mixed or C57BL/6 pure genetic backgrounds are established.
  • one half of each cohort is injected three times/week with COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c 0.2 mg/kg and 2 mg/kg COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c for the mixed and the pure C57BL/6 cohorts, respectively), while the other half of each cohort received vehicle.
  • the day following bleomycin treatment Compound selected from the table 1 ab and Table 1c is administered in dosage and root of administration selected from the Example 1.
  • the day following bleomycin treatment Compound selected from the table 1 ab and Table 1c is administered via intraperitoneal injection in the dosage 10-fold less than LD50.
  • the day following bleomycin treatment Compound selected from the table 1 ab and Table 1c is administered via intraperitoneal injection in the dosage 100-fold less than LD50.
  • the day following bleomycin treatment, Compound selected from the table 1 ab and Table 1c is administered in dosage, regimen and root of administration selected from the Example 1.
  • mice are injected with a lethal dose of isoproterenol (680 mg/kg) and the time to cardiac arrest was recorded. While 18-month-old untreated (vehicle) mice consistently show a marked acceleration of cardiac arrest compared to 12-month-old control mice, COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c -treated mice sustain youthful cardio-protection against isoproterenol, regardless of gender and genetic background. Cardio-protective signaling pathways are known to provide tolerance to metabolic stresses such as ischemia and hypoxia decline (Granfeldt et al., 2009, Cardiovasc. Res.83:234-246).
  • EXAMPLE 24 Animal studies retinopathy The breakdown of vascular beds in ischemic retinopathies—whether glycemia-driven, as in diabetic retinopathy (DR), or oxygen-driven, as in retinopathy of prematurity (ROP)—yields hypoxic/ischemic central nervous system (CNS) tissue subjected to biochemical and inflammatory stressors that compromise cellular function.
  • ischemic retinopathies whether glycemia-driven, as in diabetic retinopathy (DR), or oxygen-driven, as in retinopathy of prematurity (ROP)—yields hypoxic/ischemic central nervous system (CNS) tissue subjected to biochemical and inflammatory stressors that compromise cellular function.
  • DR diabetic retinopathy
  • ROP retinopathy of prematurity
  • Oxygen-induced retinopathy Mouse pups from different strains (C57BL/6 WT, LysM-Cre, LysM-Cre/ IRE1fl/fl, LysM-Cre/IRE1+/+, IRE1fl/fl, and LysM-Cre/ROSA26EYFPfl/fl) and their fostering mothers (CD1, Charles River) can be exposed to 75% O2 from P7 to P12 and returned to room air.
  • This model serves as a proxy to human ocular neovascular diseases such as ROP and DR characterized by a late phase of destructive pathological.
  • hypoxia-driven neovascularization develops from P14 onward
  • a therapeutic inhibition of the SASP through intravitreal delivery of the compound selected from the table 1 ab and Table 1c in mice reduces destructive retinal neovascularization in vivo.
  • a intravitreal delivery of the compound selected from the table 1 ab and Table 1c in mice reduces destructive retinal neovascularization in vivo.
  • mouse pups shall be subjected to a model of oxygen-induced retinopathy (OIR) that yields avascular neural zones similar to those observed in DR and ROP.
  • OIR oxygen-induced retinopathy
  • Mouse pups are exposed to 75% oxygen from postnatal day 7 (P7) to P12 to induce vaso-obliteration and are returned to ambient air where maximal preretinal neovascularization is reached at P17. Further details on the model can be found in Oubaha et al., Sci.
  • components of the VEGF signaling pathway are not affected.
  • Animals are treated with COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c at the dosage 50-fold less than LD50.
  • Animals are treated with COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c at the dosage 10-fold less than LD50. In yet another example, Animals are treated with COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c at the dosage 100-fold less than LD50. In yet another example, Animals are treated with COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c at the dosage selected from the Example 1. In yet another example, Animals are treated with COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c at the dosage and root of administration selected from the Example 1.
  • intravitreal injection of compound selected from the table 1 ab and Table 1c enhances vascular regeneration as assessed at P17 and suppresses pathological neovascularization.
  • administration of compound selected from the table 1 ab and Table 1c enhances vascular regeneration as assessed at P17 or/and suppresses pathological neovascularization.
  • Inclusion Criteria Nonproliferative diabetic retinopathy (DR) patients with DME. Center-involved DME with central subfield thickness (CST) ⁇ 350 ⁇ m on SD-OCT. BCVA in the study eye (most affected) of 35 Early Treatment Diabetic Retinopathy Study (ETDRS) letters or worse of 6 months duration or longer prior to screening. The measurements of efficacy are conducted by the methods known it the art.
  • at least one of the symptoms of DR is alleviated by the treatment.
  • Non-limiting examples of the symptomes are Spots or dark strings floating in the vision (floaters), Blurred vision, Fluctuating vision, Impaired color vision, Dark or empty areas in the vision, Vision loss.
  • Age-related bone loss Compound selected from the table 1 ab and Table 1c prevents age-related bone loss in mice Use of compound selected from the table 1 ab and Table 1c prevents age-related bone loss.
  • compound selected from the table 1 ab and Table 1c is treated (intraperitoneally [i.p] twice weekly) for 4 months at dosage 10-fold less than LD50.
  • compound selected from the table 1 ab and Table 1c is treated (intraperitoneally [i.p] twice weekly) for 4 months at dosage 50-fold less than LD50.
  • compound selected from the table 1 ab and Table 1c is treated (intraperitoneally [i.p] twice weekly) for 4 months at dosage 100-fold less than LD50.
  • compound selected from the table 1 ab and Table 1c is treated (intraperitoneally [i.p] twice weekly) for 4 months at dosage 500-fold less than LD50.
  • compound selected from the table 1 ab and Table 1c is treated (intraperitoneally [i.p] twice weekly) for 4 months at dosage 1000-fold less than LD50.
  • compound selected from the table 1 ab and Table 1c is treated (intraperitoneally [i.p] twice weekly) for 4 months at different dosages (0.01 mg/kg – for group #1, 0.1 mg/kg – for group #2, 1 mg/kg – for group #3, 10 mg/kg – for group #4, 25mg/kg – for group #5, 50 mg/kg – for group #6, 100 mg/kg – for group #7, 500 mg/kg – for group #8.
  • compound selected from the table 1 ab and Table 1c is treated (intraperitoneally [i.p] twice weekly) for 4 months at dosages and by the root of administration selected from the Example 1.
  • compound selected from the table 1 ab and Table 1c is treated at dosages, regimen and by the root of administration selected from the Example 1.
  • the other protocol in details is described in Farr JN, Xu M, Weivoda MM, et al. Targeting cellular senescence prevents age-related bone loss in mice [published correction appears in Nat Med.2017 Nov 7;23 (11):1384]. Nat Med.2017;23(9):1072-1079.
  • the compound, selected from the table 1 ab and Table 1c treatment results in lower p16Ink4a mRNA expression in bone relative to vehicle-treated mice as well as lower EGFP mRNA encoded by the INK-ATTAC transgene, consistent with clearance of senescent cells.
  • compound selected from the table 1 ab and Table 1c treated - versus vehicle-treated mice has better spine trabecular bone microarchitecture, with similar improvements in trabecular bone at the femur.
  • Compound selected from the table 1 ab and Table 1c treatment also results in higher cortical thickness and bone strength (by micro-finite element analysis [ ⁇ FEA]) at the femur.
  • mice are housed in ventilated cages and maintained within a pathogen-free, accredited facility under a 12- hr light/dark cycle with constant temperature (23°C) and access to food (standard mouse diet, Lab Diet 5053, St. Louis, MO) and water ad libitum. All experiments are performed in accordance with IACUC guidelines. Both sexes are studied as specified below. Genotype them to select INK-ATTACheterozygotes and aged them to 6–12 (young) or 20 (old) months. All experiments using the INK-ATTACmice are performed on transgenic line 3, which contains 13 copies of the transgene inserted into a single locus.
  • Female INK-ATTAC mice from this cohort are randomly assigned to be injected (twice weekly beginning at 20 months of age for a total of 4 months (all old mice are sacrificed at 24 months of age).
  • Young (12-month) female INK-ATTAC mice are treated using the same dosing regimen for 1.5 months (to be sacrificed at 13.5 months of age).
  • C57BL/6 male mice are obtained from the National Institute on Aging (NIA) at 7-, 20- or 22-months of age. Alternatively, C57BL/6 breeding colonies are maintained to generate animals for in vitro osteoclastogenesis assays.
  • Twenty-month-old male C57BL/6 mice are randomly assigned to once monthly treatments by oral gavage with compound selected from the table 1 ab and Table 1c or vehicle for four months5.
  • Tissue collection Mice are sacrificed, body mass is recorded and serum/plasma is collected via cardiac puncture at sacrifice and stored at ⁇ 80°C.
  • the right femur/tibia and lumbar vertebrae are fixed in 10% neutral buffered formalin and stored in ethanol to be used for micro-computed tomography ( ⁇ CT), histomorphometry, and SADS). The remaining vertebrae are used for osteocyte-enriched cell isolations.
  • ⁇ CT micro-computed tomography
  • SADS micro-computed tomography
  • the tail is collected at the base and stored in PBS soaked gauze at ⁇ 20°C for biomechanical compression and biomaterial nanoindentation testing of the caudal vertebrae.
  • PBS soaked gauze at ⁇ 20°C for in vitro analyses of osteoclast progenitors or osteoclastogenesis.
  • left femurs/tibiae are isolated and dissected free of soft tissue, epiphyses are removed, and marrow is flushed with PBS.
  • JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age.
  • perigonadal adipose tissue is collected and either fixed (for SA- ⁇ -Gal staining) or snap frozen in liquid nitrogen for RNA and subsequent rt-qPCR analyses.
  • Isolation of osteocyte-enriched cells Detailed methods and validation of our osteocyte-enriched cell isolation protocol are presented elsewhere. Briefly, mouse vertebrae are stripped of muscle/connective tissues, minced, and sequentially digested twice for 30-min in collagenase (Liberase; Roche Diagnostics GmbH, Mannheim, Germany). As shown previously, the remaining cell fraction represents a highly enriched population of osteocytes used for rt- qPCR analyses.
  • SADS analysis of senescent osteocytes Senescence-associated distention of satellites (SADS) are measured in vivo in a blinded fashion as described in the art in osteocytes located in cortical bone in the long bone diaphyses. Briefly, the decalcified right tibiae of mice are fixed, embedded in methylmethacrylate (MMA), and sectioned, followed by fluorescent in situ hybridization (FISH). Bone sections are crosslinked with 4% paraformaldehyde (PFA) for 20 min, is hed 3 times in PBS (5 min each), and dehydrated in graded ethanol as follows: 70%, 90%, and 100% (3 min each).
  • PFA paraformaldehyde
  • sections are denatured for 10 min at 80°C in hybridization buffer: 70% formamide (Sigma), 25mM MgCl2, 0.1M Tris (pH 7.2), 5% blocking reagent (Roche) containing 1.0 ⁇ g/mL of Cy3-labelled (F3002, reacts to both human and mouse), and CENPB-specific (ATTCGTTGGAAACGGGA) peptide nucleic acid (PNA) FISH probe (Panagene Inc, Korea), followed by hybridization for 2 hrs at room temperature in the dark. Slides are is hed, stained, and mounted with vectashield DAPI-containing mounting media (Life Technologies).
  • Areal bone mineral density (aBMD; g/cm2) of the lumbar spine (L1–L4) is measured by dual-energy X-ray absorptiometry (DXA) using a Lunar PIXImus densitometer (software version 1.44.005; Lunar Corp., Madison, WI). Measures of total (metaphysis), trabecular (metaphysis), and cortical (midshaft diaphysis) volumetric BMD (vBMD; mg/cm3) at the tibia are obtained using peripheral quantitative computed tomography (pQCT; Stratec XCT Research SA Plus, software v5.40, Nordland Medical Systems, Fort Atkinson, WI).
  • DXA dual-energy X-ray absorptiometry
  • Quantitative analysis of the lumbar spine (L4–L6) and distal femoral metaphysis are performed using the Viva Scan 40 ⁇ CT scanner (Scanco Medical AG, Basserdorf, Switzerland) with the following parameters: 55kVp, 145mA, high resolution, 21.5 diameter, 10.5 ⁇ m voxel size, 300 ms integration time.
  • trabecular bone mass and microarchitecture including trabecular bone volume fraction (BV/TV; %), trabecular number (Tb.N; 1/mm), trabecular thickness (Tb.Th; mm), trabecular separation (Tb.Sp; mm [higher values are associated with weaker bone]), and the structure model index (SMI), which indicates whether trabeculae are stronger, plate-like (lower values) or weaker, rod-like (higher values).
  • BV/TV trabecular bone volume fraction
  • Tb.N trabecular number
  • Tb.Th 1/mm
  • Tb.Sp trabecular separation
  • SMI structure model index
  • Cortical thickness (Ct.Th; mm) is assessed at the distal femoral metaphysis (50 slices).
  • Micro-finite element analysis ( ⁇ FEA) is performed at the femoral metaphysis to assess failure load (N; i.e., bone strength) using the manufacture’s software (Scanco Medical AG, Basserdorf, Switzerland; Finite Element-Software Version 1.13). All ⁇ CT parameters are derived using the manufacturer’s protocols. Compression loading Loading tests are performed in a blinded fashion.
  • the sixth caudal vertebrae (Ca6) is removed from the tail, stripped of soft tissue, and measured end-to-end with a digital caliper (ABSOLUTE, Mitutoyo, Aurora, IL).
  • Vertebrae are soaked in PBS for 15 to 30 minutes before testing to ensure hydration.
  • Cyanoacrylate adhesive is applied to each end of the bones to mount them between two #10–32 stainless steel nuts filled with PMMA resin.
  • the assembly is loaded into an alignment guide to impart parallel loading surfaces at each end of the bone while the adhesive cured for five minutes.
  • Compression testing is conducted with a servohydraulic test system instrumented with a 450-Newton (N) capacity load cell (Mini Bionix II, MTS Systems, Eden Prairie, MN). Bones are pre-loaded to 1 N and then loaded until failure under displacement control at a rate of 0.02 mm/s, as previously described. Force and displacement data are collected at a sample rate of 100 Hz.
  • Nanoindentation is performed in a blinded fashion.
  • the seventh caudal vertebra (Ca7) is removed from the tail and stripped of soft tissue.
  • Specimens are embedded in PMMA resin (Lecoset 100, Leco, St. Joseph, MO) and then sectioned transversely within the body of the vertebrae with a low-speed diamond saw (Isomet, Buehler, Lake Bluff, IL).
  • Sections are manually polished with a polishing/griding system (Ecomet 250, Buehler) using successively finer abrasive cloths (400, 600, 800, and 1200 grit) with a final polish using a microcloth and slurry of 0.05 ⁇ m aluminum abrasive (Union Carbide, Houston, TX).
  • Indentation testing is conducted on cortical bone with a nanoindentation system (TI 950, Hysitron, Minneapolis, MN) equipped with a diamond Berkovitch pyramidal tip. Four sites, widely distributed around cross-section, are tested on each bone. At each site, a 2 ⁇ 2 array is indented with 15 ⁇ m spacing between indents.
  • Indentation is conducted under load control at a rate of 500 ⁇ N/s to a peak load of 2000 ⁇ N with a 60 s hold before unloading to reduce viscoelastic effects, as previously described26.
  • the reduced modulus (Er; GPa), and hardness (H; GPa) are calculated using the Oliver-Pharr model, as previously described in the art. Measures are averaged over the four indents of the array to generate a value for the site. Values obtained at the four sites are averaged to generate an aggregate value for each specimen. Histomorphometric analyses All histomorphometric analyses are performed in a blinded fashion.
  • mice are injected subcutaneously with Alizarin Red (0.1mL/animal, 7.5mg/mL) and calcein (0.1 mL/animal, 2.5mg/mL) on days 9 and 2, respectively, prior to euthanasia.
  • Alizarin Red 0.1mL/animal, 7.5mg/mL
  • calcein 0.1 mL/animal, 2.5mg/mL
  • the lumbar vertebrae and right femur are isolated from female INK-ATTAC mice treated with vehicle or COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c or male C57BL/6 mice treated with vehicle or D+Q and are embedded in MMA, sectioned, and stained with Masson Trichrome to assess osteoblast numbers per bone perimeter (N.Ob/B.Pm,/mm), or stained for tartrate-resistant acid phosphatase (TRAP) activity to assess osteoclast numbers per bone perimeter, N.Oc/B.Pm,/mm).
  • sections are left unstained to quantify mineralizing surfaces (mineral apposition rate, MAR, mcm/d; bone formation rate per bone surface, BFR/BS, mcm3/mcm2/d).
  • Femurs obtained from vehicle or JAKi-treated C57BL/6 male mice are decalcified for two weeks in 12.5% EDTA, followed by paraffin embedding, sectioning, and staining with Goldner’s Trichrome stain.
  • Osteoblast (N.Ob/B.Pm) and osteoclast (N.Oc/B.Pm) numbers are assessed. The determination of bone marrow adipocyte parameters has been described in the art previously.
  • adipocyte number, adipocyte perimeter (mm), and adipocyte volume per tissue volume are measured by tracing out individual adipocytes in all the fields analyzed. All histomorphometric measurements and calculations are performed with the Osteomeasure Analysis system (Osteometrics, Atlanta, Georgia). Osteoclast differentiation and quantification Following euthanasia, long bones are isolated from C57BL/6 mice, 5–8 weeks of age. As described above, bone marrow is flushed with sterile PBS. Red blood cells (RBCs) are lysed using RBC lysis buffer (1 ⁇ ) (eBioscience, San Diego, CA), and marrow counts are obtained.
  • RBCs Red blood cells
  • Equal cell numbers are cultured overnight in vehicle (negative control), 25ng/mL rmM-CSF (positive control) (R&D Systems, Minneapolis, MN), control CM, senescent CM, or CM from senescent cells treated with the JAK inhibitor 1 (SEN/JAKi).
  • CM treatments are 1:1 with ⁇ -Minimal Essential Medium (MEM, Invitrogen, Carlsbad, CA) with 10% fetal bovine serum (FBS) and 1 ⁇ antibiotic/antimycotic.
  • Negative and positive controls are 1:1 with control RPMI 1640 used for AdMSC culture.
  • Osteoclast differentiation is carried out for two to four days, with differentiation media replaced on day three. Mature osteoclasts formed on days three and four.
  • mouse monocytes are isolated from mouse bone marrow by negative selection using the Mouse Monocyte Isolation Kit (Miltenyi Biotec, Inc., San Diego, CA) and magnetic activated cell sorting (MACS, autoMACS-Pro magnetic cell sorter, Miltenyi Biotec, Inc.). Monocytes are cultured with CON or SEN CM overnight, as described above. Non-adherent populations are collected, counted, and plated at equal cell densities in osteoclast differentiation medium.
  • RBC-lysed marrow is plated directly in osteoclast differentiation medium. Osteoclast differentiation medium is replaced on day three and osteoclast cultures are fixed on day four. Mature osteoclast cultures are fixed in 1% PFA and osteoclast differentiation is assessed by TRAP activity (Acid Phosphatase, Leukocyte Kit, Sigma, St. Louis, MO)45. Osteoclasts are defined as TRAP positive cells with greater than three nuclei.
  • MethoCult assay Bone marrow is plated in MethoCultTM GF 3434 Media (StemCell Technologies, Vancouver, BC, Canada). Each mouse is tested in duplicate.
  • Serum IL-6 and CXCL1 (IL-8) levels in JAKi-treated mice are previously measured7 using Luminex xMAP technology.
  • Mouse ELISA kits (R&D Systems) are used to measure circulating plasma levels of IL-6, CXCL1 (mouse homolog to IL-8), and PAI-1 in all the other mice. All assays are performed in a blinded fashion.
  • Sample sizes are based on pilot or previously conducted and published experiments (e.g., Syed et al.) in which statistically significant differences are observed on bone with various interventions in other laboratories. For each experiment, replicates are done. All samples presented represent biological replicates. No samples are excluded from analyses. Investigators are blinded to allocation during experiments and outcome assessments, as noted specifically above.
  • EXAMPLE 26 Cardiovascular dysfunction Effects of compound selected from the table 1 ab and Table 1c on cardiovascular function in old mice Cellular senescence is associated with cardiovascular dysfunction in humans, a major cause of morbidity and mortality in the elderly. While only mild cardiac dysfunction has been reported in old mice, substantial impairment in vascular reactivity is observed in aged mice.
  • compound selected from the table 1 ab and Table 1c is administered as a single dose, in a 5 days after an assaying cardiac function is made.
  • compound selected from the table 1 ab and Table 1c is administered by the protocol selected from the table 1 ab and Table 1c .
  • the treatment of 24-month-old mice with the compound selected from the table 1 ab and Table 1c improves left ventricular ejection fraction and fractional shortening, in some embodiments, effects that are mediated by reductions in end-systolic cardiac dimensions but not cardiac preload or alterations in cardiac mass.
  • compound selected from the table 1 ab and Table 1c yields physiologically important and consistent improvements in vascular smooth muscle sensitivity to nitroprusside.
  • compound selected from the table 1 ab and Table 1c improves cardiovascular function and reduces morbidity and mortality from cardiovascular disease.
  • EXAMPLE 27 NAFLD The incidence of non-alcoholic fatty liver disease (NAFLD) increases with age.
  • Cellular senescence refers to a state of irreversible cell-cycle arrest combined with the secretion of proinflammatory cytokines and mitochondrial dysfunction. Senescent cells contribute to age-related tissue degeneration. Any one of the Compounds selected from the table 1 ab and Table 1c reduces overall hepatic steatosis in ageing, obese and diabetic mice.
  • INK-ATTAC mice can be used at 24 months of age , treat them with compound selected from the table 1 ab and Table 1c in dosage selected from the Example 1 for 3 months. In some examples, INK-ATTAC mice can be used at 24 months of age, treat them with compound selected from the table 1 ab and Table 1c by protocol selected from the Example 1 In some examples, compound selected from the table 1 ab and Table 1c is administered to 24-month- old mice by intraperitoneal injection every 3 days, for 3 months. In some examples, vehicle or Compound selected from the table 1 ab and Table 1c are administered by oral gavage once per month for 3 months.
  • mice are injected intraperitoneally with compound selected from the table 1 ab and Table 1c or vehicle for 3 days every 2 weeks for 10 weeks in a dosage selected from the Example 1.
  • mice are killed at the age of 15 months (6 male HF (3 vehicle, 3 mice treated by the compound selected from the table 1 ab and Table 1c ), 9 female HF (4 vehicle, 5 mice treated by the compound selected from the table 1 ab and Table 1c ), 8 male control (4 vehicle, 4 mice treated by the compound selected from the table 1 ab and Table 1c ) and 5 female control (2 vehicle, 3 mice treated by the compound selected from the table 1 ab and Table 1c )).
  • the animals are killed at 27 months of age.
  • the treatments are not significantly alter body or liver weight.
  • the untreated INK-ATTAC mice at 27 months of age display higher frequencies of TAF-positive and karyomegalic hepatocytes than control AL mice at 15 months of age.
  • the compound selected from the table 1 ab and Table 1c treatment reduces the frequencies of TAF-positive hepatocytes and the average numbers of TAF per hepatocyte. In some embodiments, the total DNA damage is also reduced changed with compound selected from the table 1 ab and Table 1c .
  • the compound selected from the table 1 ab and Table 1c also reduces the percentage of karyomegalic hepatocytes.
  • the compound selected from the table 1 ab and Table 1c administration results in a reduction in hepatic fat deposition in comparison with the control.
  • Animals Mice to be fed ad libitum (AL) and dietary restricted (DR) Experiments are performed in male C57Bl/6 mice aged 3, 12 and 14.2 ⁇ 1.2 months 15 purchased from Harlan (Blackthorn, UK).
  • mice are housed in same-sex cages in groups of 4–6 (56 ⁇ 38 ⁇ 18 cm 3 , North Kent Plastics, Kent, UK) and individually identified by an ear notch. Mice are housed at 20 ⁇ 2 °C under a 12 h light/12 h dark photoperiod with lights on at 0700 hours.
  • the diet used is standard rodent pelleted chow (CRM (P); Special Diets Services, Witham, UK) for AL-fed mice and the same diet, but as smaller pellets, is offered to DR mice.
  • CCM rodent pelleted chow
  • the smaller pellet size reduced competition for food.
  • DR mice are offered 60% of AL intake (calculated based on average food intake in 90 control AL mice between 5 and 12 months of age) as one ration at 0930 hours daily.
  • mice Half of the animals are subjected to DR, while the other half, matched for body mass, food intake and age, served as AL controls. Additionally, control, young mice are killed at 3 months of age. DR is introduced at 3 months of age and lasted for 9–12 months. At the age of 12 months, some mice from the AL and DR groups had their dietary regime changed AL to DR or DR to AL for 3 months. All mice are killed at the time points mentioned above and at the end of the experiment. A variety of tissues are collected. Tissues are frozen in optimal cutting temperature compound or OCT media for cryosections, snap-frozen in liquid nitrogen for biochemistry and fixed in 10% formalin for 24 h before processing and paraffin embedding.
  • H&E Haematoxylin–eosin
  • Xpg f/ ⁇ Alb-Cre + mice in a C57BL6J/FVB F1 hybrid background; referred to as Alb- Cre
  • Alb- Cre are heterozygous for Xpg in their entire body, except for the hepatocytes in the liver, which are homozygous for Xpg after Cre excision of the floxed allele.
  • Littermates, with and without Cre-recombinase expression Xpg f/+ Alb-Cre + and Xpg f/ ⁇ Alb-Cre ⁇ respectively), are used as controls (referred to as wt).
  • mice are maintained in a controlled environment (20–22 °C, 12 h light; 12 h dark cycle) and are housed in individual ventilated cages under specific pathogen-free conditions. All animals has AL access to water and standard mouse food (CRM pellets, SDS BP Nutrition Ltd.; gross energy content 4.39 kcal g ⁇ 1 dry mass, digestible energy 3.2 kcal g ⁇ 1 ).
  • mice are killed for tissue collection.
  • Tissues are snap- frozen in liquid nitrogen, embedded in TissueTek and sliced in 10 ⁇ m thick cryosections or fixed overnight in 10% phosphate-buffered formalin, paraffin-embedded, sectioned at 3 ⁇ m and mounted on Superfrost Plus glass slides. Oil Red O and H&E images are generated using the NanoZoomer Digital slide scanner with the NDP view software (Hamamatsu Photonics, Japan). A new stock of INK-ATTAC transgenic mice is generated and genotyped as previously described . Mice are house at 2–5 mice per cage in a 12 h light/12 h dark cycle at 24 °C with free access to food (standard mouse diet, Lab Diet 5053, St Louis, MO, USA) and water in a pathogen-free facility.
  • INK-ATTAC mice are housed 2–5 per cage, at 22 ⁇ 0.5 °C on a 12–12-h day–night cycle and provided food and water AL. Mice are randomly assigned into the chow diet or HF diet group. HF food is purchased from Research Diets (cat no #D12492, 60% of calories in this diet are from fat). Db/db mice homozygotic males and females are purchased from Jackson Laboratory (Bar Harbor, ME, stock number: 000642). Mixed gender cohort consisting of 13 male db/db, 10 female db/db, 8 male db/+ and 8 female db/+ is first time treated at the age of 4 months. Animals are killed at the age of 6 months.
  • MAFs are seeded and allowed to grow for 24 h and then X- ray irradiated with 5 or 10 Gy using a PXI X-Rad 225 (RPS Services Ltd) to induce cellular senescence.
  • MAFs are treated with 100 nM of complex I inhibitor rotenone, which is replaced daily. Following 10 days of treatment, induction of senescent markers is observed.
  • Hepatocytes Hepatocytes are isolated from the livers of wild-type mice by digestion with collagenase from Clostridium histolyticum (Sigma) and then filtered through a 70- ⁇ m cell strainer. Cells are collected by centrifugation (500 r.p.m.
  • Oil Red O Preparation of Oil Red O (Sigma-Aldrich, #O1391) working solution and staining of slides is performed according to Mehlem et al. 50 and the manufacturer’s instructions. Briefly, Oil Red O working solution is prepared from stock solution mixed 3:2 with water and incubated at 4 °C for 10 min.
  • MAFs are washed briefly with PBS and fixed for 10 min with 2% paraformaldehyde dissolved in PBS.
  • DAPI solution is added for 10 min and afterwards sections are washed with PBS for 5 min.
  • Some sections are stained with ActinGreen 488 (ThermoFisher, 1 drop in 0.5 ml PBS) for 30 min and washed with PBS for 3 ⁇ 5 min.
  • 20–30 ⁇ l of Nile red/glycerol are directly added to each section, mounted on a glass microscope slide and covered with a cover slip. Images are taken immediately after mounting.
  • BODIPY 493/503 staining MAFs are washed briefly with PBS and fixed for 10 min with 2% paraformaldehyde dissolved in PBS.
  • Livers are further blocked with Avidin/Biotin (Vector Laboratories, no. SP-2001) for 15 min each. MAFs are washed briefly with PBS and fixed for 10 min with 2% paraformaldehyde dissolved in PBS. Cells are permeabilized for 45 min with PBG (0.5% BSA, 0.2% Fish Gelatine, 0.5% Triton X-100 in PBS). Primary antibodies are applied overnight at 4 °C. Slides are washed three times with PBS and incubated for 30 min with secondary antibody (no. PK-6101; Vector Lab). Antibodies are detected using a rabbit peroxidase ABC Kit (no. PK-6101; Vector Lab) according to the manufacturer’s instructions.
  • Substrate is developed using NovaRed (no. SK-4800; Vector Lab) or 3′3′- diaminobenzidine (no. SK4100, Vector Lab). Sections are counterstained with haematoxylin. For IF, sections are treated as before, and after the secondary antibody incubation, Fluorescein Avidin DCS (1:500 in PBS, no. A-2011, Vector Lab) is applied for 20 min. For IF on MAFs, Alexa Fluor secondary antibody (1:2,000; Molecular Probes) is applied for 30 min at RT. Sections or cells are stained with DAPI for 5–10 min and mounted in vectashield mounting media.
  • p21 immunohistochemistry is performed on formalin-fixed sections using rat anti-p21 antibody (clone HUGO 291H, Abcam, UK) and the ImPRESS Rat immunodetection system (MP-7444, Vector laboratories, Country) using 3′3′-diaminobenzidine (Dako, UK) as chromagen followed by counterstaining with haematoxylin. Sections are then dehydrated and coverslipped. Ten blinded consecutive non-overlapping fields are acquired at ⁇ 200 magnification and quantified as previously described More details on the protocol can be found in Ogrodnik M, Miwa S, Tchkonia T, et al. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun.
  • Glaucoma is comprised of progressive optic neuropathies characterized by degeneration of retinal ganglion cells (RGC) and resulting changes in the optic nerve. Removal of endogenous senescent retinal cells after elevated intraocular pressure (IOP) elevation by a treatment with the compound selected from the table 1 ab and Table 1c prevents loss of retinal functions and cellular structure. Compound selected from the table 1 ab and Table 1c protects retina degeneration. In one of the examples, after unilateral IOP elevation, mice are daily injected with Compound selected from the table 1 ab and Table 1c (5 mg/kg) intraperitoneally.
  • IOP intraocular pressure
  • mice are treated with compound selected from the table 1 ab and Table 1c by protocol selected from the Example 1.
  • VEP Visual evoked potential
  • Immunohistochemistry of Brn3a and activated caspase show increase of apoptosis at day 3 after IOP treatment.
  • p16 ⁇ 3MR mice are treated with Compound selected from the table 1 ab and Table 1c or vehicle for 5 days by intraperitoneal injection, similarly to the experimental procedure used for GCV.
  • p16 ⁇ 3MR mice are treated with Compound selected from the table 1 ab and Table 1c or vehicle by the protocol selected from the Example 1.
  • mice are no longer treated with drug or PBS and at day twelve after IOP elevation, functional measurement is performed and RGCs are quantified. Also in this treatment regime, Compound selected from the table 1 ab and Table 1c prevents the loss of similar to what is observed in GCV ⁇ treated animals. Additionally, VEP analysis reveals that Compound selected from the table 1 ab and Table 1c treatment with seven days “chase” still successfully prevents vision loss upon IOP elevation.
  • Animals Adult p16 ⁇ 3MR (Demaria et al., 2014) or C57BL/6 mice (12–16 weeks old, Jackson Labs) are housed in 20°C environment with standard (12 hr light/dark) cycling, food, and water available ad libitum.
  • the p16 ⁇ 3MR transgenic model in which the mice carry a trimodal reporter protein (3MR) under the control of p16 regulatory region (Demaria et al., 2014), allows potent selective removal of senescent cells.
  • the 3MR transgene encodes a fusion protein consisting of Renilla luciferase, a monomeric red fluorescent protein (mRFP) and herpes simplex virus thymidine kinase (HSV ⁇ TK) which converts ganciclovir (GCV) into a toxic DNA chain terminator to selectively kill HSV ⁇ TK expressing cells.
  • mice The experimental group of animals is treated by intraperitoneal (IP) administration of GCV (Sigma, 25 mg/kg once a day) or Compound selected from the table 1 ab and Table 1c after IOP elevation , and a control group of mice is sham ⁇ treated with PBS or vehicle (DMSO). Each mouse undergo unilateral hydrostatic pressure ⁇ induced IOP elevation to 90 mm Hg, with the contralateral eye left as an untreated control.
  • IP intraperitoneal
  • the mice are IP injected intraperitoneally with GCV or Compound selected from the table 1 ab and Table 1c at day 0 (IOP elevation day) and continued for four consecutive days. At day 5, animals are euthanized, and retinas are isolated and immunostained with anti ⁇ Brn3a antibody to evaluate the number of RGCs.
  • IOP intravenous
  • IOP intravenous
  • Stable elevated IOP 85–90 mm Hg is maintained for 60 min and controlled by IOP measurements using a veterinary rebound tonometer (Tonovet). Both eyes are lubricated throughout testing with an ophthalmic lubricant gel (GenTeal, Alcon Laboratories). Animals recovered on a Deltaphase isothermal pad (Braintree Scientific) until awake. The contralateral eye without IOP elevation served as a healthy non ⁇ IOP control (CTRL).
  • CTL non ⁇ IOP control
  • VEP measurements are taken at five days post ⁇ IOP elevation. This protocol is adapted from prior studies (Ridder & Nusinowitz, 2006). Mice are dark ⁇ adapted for at least 12 hr before the procedure. Animals are anesthetized with ketamine/xylazine and their eyes dilated as above. The top of the mouse's head is cleaned with an antiseptic solution. A scalpel is used to incise the scalp skin, and a metal electrode is inserted into the primary visual cortex through the skull, 0.8 mm deep from the cranial surface, 2.3 mm lateral to the lambda.
  • a platinum subdermal needle (Grass Telefactor) is inserted through the animal's mouth as a reference and through the tail as ground. The measurements commenced when the baseline waveform became stable, 10–15 s after attaching the electrodes. Flashes of light at 2 log cd.s/m 2 are delivered through a full ⁇ field Ganzfeld bowl at 2 Hz. Signal is amplified, digitally processed by the software (Veris Instruments), then exported, and peak ⁇ to ⁇ peak responses are analyzed in Excel (Microsoft). To isolate VEP of the measured eye from the crossed signal originating in the contralateral eye, a black aluminum foil eyepatch is placed over the eye not undergoing measurement.
  • the eyes are then dissected, the retinas flat ⁇ mounted on microscope slides, and immunostained using a standard sandwich assay with anti ⁇ Brn3a antibodies (Millipore, MAB1595) and secondary AlexaFluor 555 anti ⁇ mouse (Invitrogen, A32727).
  • Mounted samples (Fluoromount, Southern Biotech 0100 ⁇ 01) are imaged in the fluorescent microscope at 20x magnification (Biorevo BZ ⁇ X700, Keyence), focusing on the central retina surrounding the optic nerve.
  • Overall damage and retina morphology are also taken into consideration for optimal assessment of the retina integrity.
  • Micrographs are quantified using manufacturer software for Brn3a ⁇ positive cells in 6 independent 350 ⁇ 350 ⁇ m areas per flat mount.
  • RNA extraction from mouse tissues, cDNA synthesis, and RT ⁇ qPCR experiments are performed as previously described (Skowronska ⁇ Krawczyk et al., 2015). Assays are performed in triplicate. Relative mRNA levels are calculated by normalizing results using GAPDH. The primers used for RT ⁇ qPCR are the same as in (Skowronska ⁇ Krawczyk et al., 2015). The differences in quantitative PCR data are analyzed with an independent two ⁇ sample t test. SA ⁇ gal assay to test senescence on retinas mouse eyes Senescence assays are performed using the Senescence b ⁇ Galactosidase Staining Kit (Cell Signaling) according to the manufacturer's protocol.
  • RNA ⁇ Seq analysis High ⁇ quality RNA is extracted using TRIzol Reagent (Invitrogen) and treated with TURBO DNA ⁇ free Kit (Invitrogen).
  • RNA ⁇ Seq libraries are made from 1 ⁇ g total RNA per tissue sample using TruSeq stranded mRNA Library Prep Kit (Illumina, kit cat. no.20020597) according to the manufacturer's instructions. The libraries are size ⁇ selected using Agencourt Ampure XP beads (Beckman Coulter) and quality ⁇ checked by Bioanalyzer (Agilent).
  • RNA ⁇ Seq paired ⁇ end reads sequence data (PE50: 2 ⁇ 50 bp) are obtained on a HiSeq4000. RNA ⁇ Seq reads are counted using HOMER software considering only exonic regions for RefSeq genes. More details on the protocol can be found in Rocha LR, Nguyen Huu VA, Palomino La Torre C, et al. Early removal of senescent cells protects retinal ganglion cells loss in experimental ocular hypertension. Aging Cell.2020;19(2):e13089.
  • Wild-type and PS19 mice are treated with a repeating schedule of COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c , beginning at weaning age and continuing until the mice reached six months of age. Notably, this treatment prevents the upregulation of senescence-associated genes and attenuates tau phosphorylation in PS19 mice.
  • mice are treated with compound selected from the table 1 ab and Table 1c by protocol selected from the Example 1.
  • MAPT P301S PS19 mouse model of tau-dependent neurodegenerative disease accumulates p16 INK4A - positive senescent astrocytes and microglia.
  • clearance of these cells as they arise using compound selected from the table 1 ab and Table 1c prevents gliosis, hyperphosphorylation of both soluble and insoluble tau leading to neurofibrillary tangle deposition, and degeneration of cortical and hippocampal neurons, thus preserving cognitive function
  • Pharmacological intervention with a Compound selected from the table 1 ab and Table 1c modulates tau aggregation.
  • PS19;ATTAC mice Compared with control mice, vehicle-treated PS19;ATTAC mice has nearly double the number of cells containing X-Gal crystals in both the hippocampus and the cortex, whereas COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c -treated PS19;ATTAC mice has a similar incidence of X-Gal crystals as control mice.
  • a distinguishing characteristic of PS19 mice is the development of aggregates consisting of hyperphosphorylated tau protein by six months of age. One can measure the levels of soluble total and phosphorylated tau (S202/T205) in addition to the level of insoluble phosphorylated tau in vehicle-treated PS19;ATTAC and COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c -treated ATTAC and PS19;ATTAC mice.
  • Vehicle-treated PS19;ATTAC mice displays increased levels both of soluble total and phosphorylated tau and of insoluble phosphorylated tau.
  • Compound selected from the table 1 ab and Table 1c -treated PS19;ATTAC mice has identical levels of soluble total tau to vehicle-treated PS19;ATTAC mice, indicating that overexpression of tau from the transgene is maintained.
  • Treatment of PS19;ATTAC mice with Compound selected from the table 1 ab and Table 1c -treated reduces the amount of phosphorylated tau in both the soluble and the insoluble fractions.
  • NFT deposition is attenuated upon treatment with Compound selected from the table 1 ab and Table 1c -treated in both the cortex and the hippocampus of PS19;ATTAC mice, one can perform assessments for degeneration in these areas.
  • the overt brain size of vehicle-treated PS19;ATTAC mice is reduced compared to both ATTAC and Compound selected from the table 1 ab and Table 1c -treated PS19;ATTAC mice.
  • the administration of Compound selected from the table 1 ab and Table 1c prevents thinning of the dentate gyrus and increased neuron density.
  • Wild-type and PS19 mice are treated with a repeating schedule of COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c , beginning at weaning age and continuing until the mice reached six months of age.
  • this treatment prevents the upregulation of senescence-associated genes and attenuated tau phosphorylation in PS19 mice.
  • the continuous clearance of p16 In 4a -expressing senescent cells by Compound selected from the table 1 ab and Table 1c before disease onset in a model of aggressive tauopathy has a treatment effect on various aspects of disease progression, including gliosis, NFT formation, neurodegeneration and cognitive decline.
  • Senescent-cell clearance by Compound selected from the table 1 ab and Table 1c also has a notable effect on the accumulation of phosphorylated tau protein in both the soluble and insoluble fractions.
  • the amount of total soluble tau is unchanged in Compound selected from the table 1 ab and Table 1c -treated PS19;ATTAC mice, indicating that the aberrant hyperphosphorylation of tau protein and subsequent aggregation into NFTs is mediated by extracellular signalling from p16 In k4a -expressing senescent glial cells.
  • the absence of neurodegeneration in mice treated with Compound selected from the table 1 ab and Table 1c -treated demonstrates that the attenuation of disease severity does not result from the clearance of neurons containing NFTs.
  • COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c can modulate senescent cells and attenuate tau phosphorylation.
  • a RT–qPCR analysis of the expression of senescence markers from the hippocampus (left) and the cortex (right) of six-month- old mice treated with either vehicle (–COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c ) or COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c (+COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c ). normalized to the WT – COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c group Mouse strains and drug treatment.
  • MAPT P301S PS19 (PS19) mice are purchased from The Jackson Laboratory (stock no. 008169) and bred to C57BL/6 for three generations. C57BL/6 ATTAC transgenic mice are as previously described. Male PS19 mice are bred to ATTAC females to generate cohorts of ATTAC and PS19;ATTAC mice. All mice are on a pure C57BL/6 genetic background. Mice from this cohort are randomly assigned to receive COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c (AP; B/B homod- imerizer; Clontech) or vehicle twice a week beginning at weaning age (3 weeks).
  • Compound selected from the table 1 ab and Table 1c pulse- treated mice received a dose for five consecutive days before tissue collection.
  • the intervention by Compound selected from the table 1 ab and Table 1c is performed in C57BL/6 wild-type and PS19 mice.
  • mice are assigned to receive either COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c or vehicle (Phosal 50 PG, Lipoid NC0130871, 60%; PEG400, Sigma 91893, 30%; ethanol, 10%).
  • COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c is administered on a repeating regimen of five consecutive days of treatment followed by 16 days of rest.
  • mice are housed in a 12 h:12 h light:dark cycle environment in pathogen-free barrier conditions as previously described in detail. Compliance with relevant ethical regulations and all animal procedures are reviewed and approved by the Mayo Clinic Institutional Animal Care and Use Committee. Statistical analysis. Prism software is used for all statistical analysis. A Student’s two-tailed unpaired t-test with Welch’s correction is used in 1 ab and Table 1c and Extended Data 4b–e; two-way ANOVA with Tukey’s multiple comparisons test is used for 3d and Extended Data 10; and one-way ANOVA with Tukey’s multiple comparisons test is used in all other figures.
  • mice are transcardially perfused with ice-cold Dulbecco’s phosphate-buffered saline (DPBS; pH 7.4) until fluid run- off is clear. This is followed by perfusion with 4% para- formaldehyde (PFA) for 10 min at a rate of 3 ml min ⁇ 1 , and then ice-cold DPBS is perfused again for 2 min at the same rate to remove the remaining fixative. Brains are then isolated and the hippocampus and cortex are dissected out.
  • DPBS phosphate-buffered saline
  • Two grids from each tissue are produced, and >100 cells are scanned per grid at a magnification of 20,000 ⁇ to detect cells containing X- Gal crystals. On average, half of all cells examined are neurons. Cells with one or more crystals, and the total number of cells, are counted. Cells containing crystals are imaged and independently assessed for distinguishing morphology. To define cell type, the following criteria are applied. Astrocytes: circular nucleus with spattered electron density pattern; microglia: abnormally shaped nucleus with a much darker, often phagosome-containing cytoplasm; and neuron: large circular nucleus with less electron density and periodically denoted by an offshooting axon.
  • Novel-object recognition testing is performed as previously described. In brief, mice from each cohort are acclimatized to a 50 cm ⁇ 50 cm testing environment for a period of two minutes. After acclimatization, the mice are removed, the testing area is cleaned with 70% ethanol, and two identical scented candles are placed in either corner of the testing area approx- imately 5 cm from either wall. Mice are reintroduced, and the ratio of both the number of visits and the time spent at each candle is recorded for a period of ten minutes.
  • RNA extraction, cDNA synthesis and RT–qPCR analysis are performed on hippocampi and cortical samples from mouse brains as previously described.
  • mice are transcar- dially perfused as described in ‘Senescence-associated ⁇ -galactosidase transmission electronmicroscopy(Gal- TEM)’.Brainsarestoredin4%PFAovernightat4°C and then cryoprotected by incubating in a 30% sucrose solution for 48 h at 4 °C. Samples are sectioned into 30- ⁇ M-thick coronal sections and stored in antifreeze solution (300 g sucrose, 300 ml ethylene glycol, 500 ml PBS) at ⁇ 20 °C.
  • Nissl staining (bregma ⁇ 2.1 to ⁇ 2.4 mm), thioflavin S staining (bregma ⁇ 1.4 to ⁇ 1.6 mm), and phospho-tau S202/S205 (Thermo Fisher, MN1020; 1:500), phospho-tau T231 (Thermo Fisher, MN1040; 1:500), phospho-tau S396 (Abcam, 109390; 1:500), and GFAP (Dako, Z0334; 1:500) and IBA1 (Novus, NB100-1028; 1:100) immunohisto- chemistry staining (bregma 1.6 to 1.0 mm and lateral 2.0 to 2.7 mm) is performed on free-floating sections as described.
  • mice are injected with EdU (Carbosynth, NE08701; 75 mg kg ⁇ 1 ) intraperitoneally 24 h before euthanasia. Imaging of EdU-positive cells (lateral 0.75 to 1.25 mm) is performed following the manufacturer’s instructions (Invitrogen Click-iT EdU Alexa Fluor 488 Imaging Kit, C10337).
  • IBA1 (Wako, 019-19741; 1:500) immuno- fluorescent staining and IBA1/EdU colocalization assessments are performed as previously described 28 .
  • TUNEL staining (lateral 0.75 to 1.25 mm) is performed according to the manufacturer’s instructions (Roche In situ Cell Death Detection Kit, Fluorescein: 11684795910).
  • Thioflavin S, EdU/IBA1 colocalization, and in vivo TUNEL-stained images are acquired on a Zeiss LSM 780 confocal system using multi-track configuration. Single-cell preparation and FACS.
  • Dissociation of cerebral tissue is performed using the Adult Brain Dissociation kit from Miltenyi (MACS, 130-107-677), according to the manufacturer’s instructions.
  • IncuCyte tracking of basal and activated astrocytes To track basal and activated astrocyticresponsetoAP,astrocytesareplatedina48-wellcultureplate(10,000cells per well) and placed into the IncuCyte S3 Live-Cell Analysis System.
  • the IncuCyte System is a time-lapse imaging system that records changes in cell culture through photographic capture of the culture well within the incubator.
  • Cultures are acclimatized to the system for a period of 6 h, then exposed to medium containing IFN ⁇ (R&D Systems, 285-IF; 200 ng ml ⁇ 1 ), LPS (Sigma, L2654; 100 ng ml ⁇ 1 ) or a combination of both for a period of 24 h to induce an inflammatory response 35 .
  • Cells are also plated on 10-well slides and processed in tandem for immunofluo- rescence staining to verify activation status with anti-GFAP (DAKO, Z0334; 1:500) and counterstained with DAPI (Invitrogen, D1306; 1:1,000).
  • phase confluency difference is calculated by subtracting the final phase confluency of each image from its initial value. More details on the protocol can be found in Bussian, T.J., Aziz, A., Meyer, C.F. et al.
  • CIA-induced DBA/1J mice are treated with compound selected from the table 1 ab and Table 1c or methotrexate (MTX) by protocol selected from the Example 1.
  • Arthritis severity is evaluated by arthritic score and joint histopathology. Draining lymph node (dLN), joint and peripheral-blood mononuclear-cell (PBMC) counts, and activation/localization of T-/B-lymphocytes, dendritic cells (DCs) and neutrophils are examined by FACS analysis. Serum anti-type-II-collagen-(CII) antibody levels and cultured-splenocyte and serum cytokines are also evaluated.
  • Compound selected from the table 1 ab and Table 1c reduces CIA-induced arthritic score, histopathology and leukocyte counts. Besides, compound selected from the table 1 ab and Table 1c suppresses CIA-activated T-cells including CD3+, CD3+/CD69+, CD8+, CD4+ and CD4+/CD25+ in dLNs or joints. It also modifies CD19+ or CD20+/CD23+ (B-cells), MHCII+/CD11c+ (DCs) and Gr-1+/CD11b+ (neutrophil) subpopulations. It further depresses total anti-CII-IgG, anti-CII-IgG1 and anti-CII-IgG2a antibody productions.
  • compound selected from the table 1 ab and Table 1c suppresses CIA-induced serum TNF- ⁇ , IL-1 ⁇ and IL-6 levels.
  • Compound selected from the table 1 ab and Table 1c also blockes IFN- ⁇ , IL-17 and IL-6 cytokines while it does not affect IL-10 but enhances IL-4 in splenocytes.
  • Compound selected from the table 1 ab and Table 1c have immunomodulatory and anti-inflammatory properties with the capacity to ameliorate the inflammatory response in CIA-mice.
  • compound selected from the table 1 ab and Table 1c reduces severity of arthritis in CIA mice.
  • DBA1J mice with established CIA are injected either with phosphate-buffered saline (control) or 0.2 mg/kg methotrexate intraperitoneally or with oral dose of compound selected from the table 1 ab and Table 1c 30 or 90 mg/kg daily for 5 weeks after day 29 post- immunization.
  • compound selected from the table 1 ab and Table 1c reduces severity of arthritis in CIA mice.
  • DBA1J mice with established CIA are injected either with phosphate-buffered saline (control) or 0.2 mg/kg methotrexate intraperitoneally or are adminiterested the compound selected from the table 1 ab and Table 1c daily for 5 weeks after day 29 post-immunization by the root of administration and dosage selected from the Example 1.
  • compound selected from the table 1 ab and Table 1c reduces severity of arthritis in CIA mice.
  • DBA1J mice with established CIA are injected either with phosphate-buffered saline (control) or 0.2 mg/kg methotrexate intraperitoneally or are adminiterested the compound selected from the table 1 ab and Table 1c after day 29 post-immunization by the protocol selected from the Example 1.
  • Arthritic score is assessed for the indicated weeks after the onset of arthritis. Paw joints are obtained at the end of the experiment as indicated in the Materials and methods section. Joint tissues should be formalin-fixed, decalcified, sectioned, and stained with hematoxylin-eosin stains and analyzed as na ⁇ ve unimmunized, vehicle CIA-induced, 30 mg/kg compound selected from the table 1 ab and Table 1c (or, in other example - other dosage suggested in such other example) or 0.2 mg/kg MTX, treated arthritic animals. Extensive leukocyte infiltration and synovial expansion into the articular surface are observed in vehicle-treated animals.
  • Paw joints of CIA-mice demonstrates extensive inflammatory cell infiltration, synovial hyperplasia, cartilage and bone erosions.
  • “Compound selected from the table 1 ab and Table 1c ” recipients developes a less cellular infiltration with smooth articular surface and wider articular space , lesser cartilage erosion or bone damage, which is comparable to that of na ⁇ ve or MTX groups.
  • CIA induction is performed in accordance with nature protocol (Brand DD, Latham KA, Rosloniec EF.
  • bovine collagen type II (Chondrex, Inc., WA, USA)
  • CFA complete Freund's adjuvant
  • mice are given booster doses of 200 ⁇ l of CII in incomplete Freund's adjuvant (IFA).
  • IFA incomplete Freund's adjuvant
  • Animals Male DBA/1J mice (8-weeks old and 20–22 g weight) are obtained from (The Jackson Laboratory; Bar Harbor, Maine, USA), and housed under specific pathogen free conditions with free access to feed and water. Mice are acclimated for two weeks before use and all in vivo experiments are carried out in a specific pathogen-free facility. The experiments are conducted according to internationally accepted guidelines on the use of laboratory animals. Collagen-induced arthritis CIA induction is performed in accordance with nature protocol known in the art.
  • bovine collagen type II (Chondrex, Inc., WA, USA) is dissolved in 10 mM acetic acid to 2 mg/ml. This solution is then emulsified in equal volumes of complete Freund's adjuvant (CFA) containing 250 ⁇ g/mouse heat-killed mycobacterium tubercu- losis H37Ra (Difco laboratories Inc., MI, USA). Then, 200 ⁇ l of the emul- sion is injected intradermally at the base of the tail of each mouse on day 0. At day 21 after primary immunization, mice are given booster doses of 200 ⁇ l of CII in incomplete Freund's adjuvant (IFA).
  • CFA complete Freund's adjuvant
  • mice are randomly allotted into five groups of 10 each.
  • Group 1 is used as the non-immunized (normal), whereas mice in Groups 2–5 are immunized on day 0.
  • Group 2 is immunized and untreated vehicle [phosphate-buffered saline (PBS) once daily, orally];
  • Group 3 is treated with dose of compound selected from the table 1 ab and Table 1c (90 mg/kg/day);
  • Group 4 is treated with low dose of compound selected from the table 1 ab and Table 1c (30 mg/kg/day) both doses are given by oral gavage; and
  • Group 5 is treated with methotrexate (MTX 0.2 mg/kg/day, sc.).
  • PBS phosphate-buffered saline
  • Compound selected from the table 1 ab and Table 1c treatment is initiated at 8 days after booster dose and given orally 5 days/week for 5 weeks.
  • the compound selected from the table 1 ab and Table 1c is administered by protocol suggested in Example 1.
  • Clinical assessment of arthritis The clinical arthritis is assessed daily beginning from 18 days of primary immunization and arthritic score is conducted by three in- dependent, blinded examiners for 3 times per week.
  • PBMCs peripheral blood mononuclear cells
  • the cells are ished, counted, and incubated with fluorochrome-conjugated antibodies for fluorescence-activated cell sorter (FACS) analysis.
  • FACS fluorescence-activated cell sorter
  • the cells are harvested, ished, and incubated with antibodies for FACS analysis (FACSCalibur; BD Biosciences, San Jose, CA). Characterization of specific subpopulations, developmental stages, and activation status of cells from lymph node or joint is performed by multiparameter FACS analysis.
  • the T- or B-lymphocyte subpopulations, neutrophils and antigen presenting cells are sorted using cell surface marker antibodies. Antibodies from eBioscience, Inc.
  • anti-CD3-fluorescein isothiocyanate FITC
  • anti-CD4-Alexa Fluor 488 anti-B20-FITC
  • anti-CD8-phycoerythrin PE
  • anti-CD4-PE anti-CD25-allophycocyanin
  • anti-CD19-PE anti-Gr-1-PE
  • anti-CD11b-FITC anti-MHC-II-PE
  • anti-CD11c-APC anti-CD69-APC.
  • mice are bled at the termination of experiment, and sera are analyzed for anti-CII total IgG, IgG1 and IgG2a antibody levels by quan- titative ELISA. Briefly, enzyme-linked immunosorbent assay (ELISA) plates (Thermo Fisher Scientific, NY, USA) are coated with 10 ⁇ g/ml of type II collagen dissolved in Tris buffer (50 M Tris, containing 200 mM NaCl, pH 7.4, 0.1% Tween 20), ished and blocked with 3% bovine serum albumin in Tris buffer, and then incubated with se- rial dilutions of test sera overnight at 4 °C.
  • ELISA enzyme-linked immunosorbent assay
  • IgG, IgG1 or IgG2a is detected by incubation for 1 h with horseradish peroxidase-conjugated sheep anti-mouse IgG (BD Bio- sciences). After ishing, plates are developed using 2,2′-azino-bis 3-ethylbenzothiazoline-6-sulphonic acid (ABTS) (Roche Diagnostic Systems, IN, USA) as substrate, the reaction is stopp, and the absorbance is then measured at 450 nm in a Spec- tra Max Plus reader (R & D systems, Minneapolis, MN, USA).
  • ABTS 2,2′-azino-bis 3-ethylbenzothiazoline-6-sulphonic acid
  • a standard serum from arthritic and nonimmunized syngeneic mice is added to each plate in serial dilutions as positive and negative controls, respectively.
  • Preparation of spleen cell suspensions from CIA mice Spleens are aseptically removed from 5 mice in each group and ished with cold PBS and the tissues are minced. Single-cell suspensions are prepared by passing each spleen through a 70- ⁇ m-cell strainer with the end of a 10-ml plastic syringe plunger (BD FalconTM; Bedford, MA, USA). The suspensions are then centrifuged on Ficoll- Paque TM at 1200 g for 5 min (Fisher Scientific, PA, USA) and the cell layers are collected.
  • the cells are resuspended in triplicate in 24-well plates cultured with RPMI 1640 medium containing 50 mM 2- mercaptoethanol, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin, 50 ⁇ g/ml gentamicin and 10% fetal bovine serum (FBS).
  • the cells are dispersed in trypsin, counted and adjusted to 2 ⁇ cells/ml.
  • immunoassays are performed using antibodies against IL-1 ⁇ , TNF- ⁇ , IL-6, IFN- ⁇ , IL-17 and IL-10 of a mouse cytokine immunoassay kit (R&D Systems, MN, USA). Blood samples are collected from all mice via cardiac punc- tures prior to sacrifice and allowed to clot for 2 h at room temperature, and centrifuged at 2000 g for 20 min at 4 °C to obtain serum.
  • splenocyte cytokine levels single-cell suspensions at a density of 2 ⁇ 1cells/well are cultured in triplicate to 24-well plates. After 24 h incubation, splenocytes in the absence or presence of compound selected from the table 1 ab and Table 1c (30 mg/kg) or MTX (0.2 mg/kg) are stimulated with 50 ⁇ g/ml anti-collagen type II peptide (Cyanogen bromide cleaved peptide 11 (CB11) of bovine type II collagen (Chondrex, Inc., WA, USA)), for 48 h at 37 °C in a 5% CO2-humidified atmosphere.
  • compound selected from the table 1 ab and Table 1c (30 mg/kg) or MTX (0.2 mg/kg) are stimulated with 50 ⁇ g/ml anti-collagen type II peptide (Cyanogen bromide cleaved peptide 11 (CB11) of bovine type II collagen (Chondrex, Inc
  • Cy- tokine (IL-17, IL-6, INF- ⁇ , IL-10 and IL-4) levels in the culture media supernatants are assayed as indicated above in accordance with the manufacturer's recommendations.
  • Histological analysis For histologic analysis, hind paws are randomly collected from 5 mice of each group, fixed in 10% buffered formalin and decalcified in 15% EDTA. The paws are processed for paraffin embedding with 5- ⁇ m sagittal serial tissue sections of whole hind paws and stained with hematoxylin and eosin (H&E) according to standard methods.
  • H&E hematoxylin and eosin
  • the systemic arthritis is induced by the injection of bCII and CFA at day 0, plus a booster immunization on day 21 in DBA/1J mice.
  • the treatment by compound selected from the table 1 ab and Table 1c is initiated after 8 days of booster dose and assessed the severity of the arthritis with an established arthritic score system.
  • the arthritic scores indicate that arthritis is progressed rapidly in vehicle-treated mice and the maximal clinical symptoms are achieved at 3 weeks after the onset of arthritis.
  • Compound selected from the table 1 ab and Table 1c shows a decrease in the severity of paw swelling and reduction in arthritis score and this pharmacological action appeares to be dose-dependent.
  • the arthritic scores of the mice treated with high (90)- and low (30 mg/kg) doses of compound selected from the table 1 ab and Table 1c are 60–70 and 52–59% decrease in the maximal arthritic scores.
  • MTX 0.2 mg/kg
  • Histology of the severity of joint inflammation and cartilage destruction CIA and RA are well characterized with synovial hyperplasia, synovitis, pannus formation, and cartilage and bone erosion in the joint.
  • histological analysis is carried out on the mouse hind paws. Consistent with the trend toward reduced paw swelling, there is notably a decreased histological signs of inflammation, and cartilage and bone destruction.
  • the vehicle treatment group had serious cellular infiltra- tion, synovial hyperplasia, cartilage damage and bone erosion with a high cumulative histological score, whereas significantly diminished signs of the above indicated pathological lesions, are observed in compound selected from the table 1 ab and Table 1c - and MTX-treated groups compared to vehicle group. This suggests that, in addition to the reduction in inflammatory infiltration, compound selected from the table 1 ab and Table 1c administration could attenuate the inflammatory response of infiltrating/proliferating synovial cells. 3.3.
  • CD4 and CD8 T-lymphocyte proliferation are increased in dLN of CIA mice with the higher proportion of CD4 T- lymphocytes. While the decrease in CD8 T-cells is significant in compound selected from the table 1 ab and Table 1c or MTX treated groups, the difference in CD4 T-cell subpopulations is not statistically significant. Similarly, CIA-induced CD4/ CD25 double positive (black bars) and CD3/CD69 double positive (early T-cell activation marker, gray bar) subpopulations are markedly reduced by compound selected from the table 1 ab and Table 1c or MTX treatment .
  • CIA-induced B-lymphocyte activation (increase in CD20/ CD23 double positive cell sub-population) is reversed to normal level by compound selected from the table 1 ab and Table 1c or MTX treatment .
  • co-localization of CIA-activated CD3 expressing T- lymphocytes and Gr-1/CD11b expressing neutrophils in joints are highly diminished in compound selected from the table 1 ab and Table 1c or MTX groups, suggesting the inhibitory ef- fect of compound selected from the table 1 ab and Table 1c in leukocyte infiltration to the synovium, which indicates its beneficial therapeutic property.
  • CIA-induced upregulation of MHCII/CD11c expressing antigen presenting cells (APCs) in the dLN is highly reduced in compound selected from the table 1 ab and Table 1c or MTX treated CIA-mice .
  • Compound selected from the table 1 ab and Table 1c treatment inhibits humoral collagen-specific immunity
  • the activated dendritic cells (DCs) migrate to dLNs and present the processed CII peptide on appropriate MHC class II molecules to na ⁇ ve T cells. Engagement of the activated DCs and T-cells creates an environment in which adaptive immunity to CII is induced.
  • the compound selected from the table 1 ab and Table 1c treatment resulted in a significant suppression of anti-CII autoantibody production.
  • the levels of anti-bCII total IgG, IgG1 and IgG2a are reduced in compound selected from the table 1 ab and Table 1c or MTX-treated groups.
  • the result suggests that compound selected from the table 1 ab and Table 1c treatment of mice with chronic arthritis resulted in a reduction in the serum levels of anti-CII IgG antibodies, which correlates with its effect in activated B-cells .
  • Compound selected from the table 1 ab and Table 1c modulates serum cytokine levels in CIA mice
  • Levels of pro-inflammatory and anti-inflammatory cytokines in serum are analyzed using a multiplex immunoassay on day 48 after immunization with CIA. Consistent with the joint swelling, TNF- ⁇ , IL-1 ⁇ , IL-6, and IL-17 in the vehicle-treated CIA mice are systemically over-produced in serum.
  • the elevated cytokine levels in mice treated with compound selected from the table 1 ab and Table 1c are decreased progressively in a manner correlated positively with the degree of joint swelling in individual animals. In the model, the raised serum IL-1 ⁇ levels in CIA mice are markedly increased.
  • Compound selected from the table 1 ab and Table 1c influences the balance of cytokines in CIA-derived cultured and anti-CII peptide-treated splenocytes Since an imbalance between pro- and anti-inflammatory cytokine activities favors the induction of autoimmunity, chronic inflammation and thereby joint damage, one, in addition to serum levels also, can examine whether such an imbalance could be modulated by compound selected from the table 1 ab and Table 1c treatment in cultured and C-II peptide stimulated-splenocytes isolated from CIA mice. In some examples, the compound selected from the table 1 ab and Table 1c decreases the production of proinflammatory cytokines including Il-17, IL-6 and IFN- ⁇ .
  • compound selected from the table 1 ab and Table 1c promotes the anti- inflammatory cytokine, IL-4 production while it does not affect the level of another anti-inflammatory cytokine, IL-10.
  • compound selected from the table 1 ab and Table 1c may modulate the balance of cytokines within a complex regulatory network related to specific immunological pro-cesses that promote autoimmunity, chronic inflammation and tissue destruction. More Details on the protocol can be found in Endale M, Lee WM, Kwak YS, Kim NM, Kim BK, Kim SH, Cho J, Kim S, Park SC, Yun BS, Ko D, Rhee M. Torilin ameliorates type II collagen-induced arthritis in mouse model of rheumatoid arthritis.
  • Compound selected from the table 1 ab and Table 1c effectively decreases HDF senescence induced by HGPS, chronological aging, ultraviolet- B radiation, and etoposide treatment, without inducing significant cell death, and likely by modulating longevity and senescence pathways.
  • UVB Ultraviolet-B
  • UVB doses of 0.05 J/cm2 and 0.1 J/cm2 effectively induce dose-dependent cellular senescence, as can be verified by SA-BGal staining relative to cell count, and reduces overall cell count).
  • UVB- exposed fibroblasts from 30 and 79 year old donors treated with increasing concentrations of Compound selected from the table 1 ab and Table 1c present decreases SA-BGal staining/nuclei and prevents UVB-induced cell death.
  • To support the reduction in senescence one can measure gene expression of cells exposed to 0.1 J/cm2 and treated with Compound selected from the table 1 ab and Table 1c .
  • UVB-damaged 30yr fibroblasts treated with Compound selected from the table 1 ab and Table 1c displays a significant decrease in the mRNA expression of genes related to aging, inflammation and SASP (B2M, IL-6, and IL-8), and a trend towards a decrease in senescence (P16).
  • UVB-dosed 79yr HDFs treated with Compound selected from the table 1 ab and Table 1c a trend is observed for decreased gene expression of P16 and B2M but no change in inflammatory markers is observed with Compound selected from the table 1 ab and Table 1c treatment.
  • Compound selected from the table 1 ab and Table 1c also decreases the levels of the senescence marker P21 in 30yr and 79yr HDFs after treatment with an acute dose of UVB 2E.
  • Compound selected from the table 1 ab and Table 1c reduces biological age in skin models.
  • In order to assess whether Compound selected from the table 1 ab and Table 1c treatment would effectively decrease cellular senescence in more complex and representative skin samples one can replicate skin aging in vitro by building 3D skin equivalents using fibroblast and keratinocyte cultures derived from elderly donors. The aged phenotype can be confirmed by measuring the overall structure and quality of the skin, which decreased according to the donor age.
  • the effect of both molecules is quantified according to morphological changes analyzed through an unbiased skin score and shows that, while Compound selected from the table 1 ab and Table 1c treatment increases the score, Rapamycin treatment lead to a decrease.
  • the mRNA expression of the Compound selected from the table 1 ab and Table 1c -treated epidermis from 3D skin equivalents built from donors aged 32, 48, and 60 years exhibits a significant decrease in P16 and a trend towards decreases IL6, in addition to a significant increase in Keratin 1 (a marker of keratinocyte terminal differentiation) and 14 (a marker of non-differentiated, proliferative keratinocytes).
  • Rapamycin induces a significant increase in P16 expression, a trend towards increased expression of inflammatory markers (IL6 and IL8), and a significant decrease in Keratin 1 gene expression levels 3B).
  • Compound selected from the table 1 ab and Table 1c treatment promotes a significant reduction in B2M gene expression, a pro-aging factor, as well as in the expression ofIL8.
  • Rapamycin treatment induces no significant changes in these markers and increases Matrix Metalloproteinase-1 (MMP1) gene expression, indicative of breakdown of the extracellular matrix 3C).
  • Rapamycin and Compound selected from the table 1 ab and Table 1c reduce gene expression of Hyaluronidase-1 (HYAL1), which plays a role in the degradation of hyaluronic acid.
  • HYAL1 Hyaluronidase-1
  • Compound selected from the table 1 ab and Table 1c treatment stimulates increased gene expression of Collagen 1 and Hyaluronan Synthase-2 (HAS2) 3C). Therefore, compared to Rapamycin, Compound selected from the table 1 ab and Table 1c treatment of 3D human skin equivalents improvs numerous markers of skin health and longevity.
  • HAS2 Hyaluronan Synthase-2
  • Compound selected from the table 1 ab and Table 1c treatment of 3D human skin equivalents improvs numerous markers of skin health and longevity.
  • Histological imaging can show an increase in epidermal area with increasing concentration of Compound selected from the table 1 ab and Table 1c and no significant changes are observed with Rapamycin treatment compared to control.
  • both Compound selected from the table 1 ab and Table 1c and Rapamycin decreases P16 mRNA expression yet only Compound selected from the table 1 ab and Table 1c treatments decreases B2M expression.
  • IL8 and tyrosinase (TYR) gene expression are reduced in all treatments though an increase in Keratin 1 and 14 is only observed with Compound selected from the table 1 ab and Table 1c treatment.
  • Compound selected from the table 1 ab and Table 1c treatment more drastically decreased P16 mRNA expression compared to Rapamycin treatment and only the higher concentration of Compound selected from the table 1 ab and Table 1c is able to reduce B2M gene expression.
  • Rapamycin treatment reduces both IL8 and HYAL1 gene expression and, similarly to Compound selected from the table 1 ab and Table 1c , is able to increase HAS2 expression.
  • Compound selected from the table 1 ab and Table 1c treatment also increases dermal Collagen 1 and the proliferation marker Ki67 in dermis though does increase HYAL1 and MMP1 gene expression in the higher concentration 3G).
  • the 79 year old ex vivo skin biopsies are processed for DNA isolation and methylome analysis. Using the Skin- Specific DNA methylation algorithm (Boroni, M. et al.
  • the cells can be purchased from Coriell Institute for Medical Research included HDF 71 yr (AG05811, XX, arm, caucasian), HDF 84 yr (AG11725, XX, arm, caucasian) and HDF 90 yr (AG08712, XX, arm, caucasian).
  • Cells can be purchased from MatTek Life Science included HDF 60 yr (F13400A, XX, african-american), keratinocytes 60yr (K13400A, XX, african-american), neonatal HDFs (F90800, XY, foreskin, caucasian), neonatal keratinocytes (K90800A, XY,foreskin, caucasian).
  • All other cells are isolated from ex vivo human skin obtained from ZenBio (Research Triangle, NC). All skin samples are from XX donors, caucasian, and abdomen area. The ages included 30, 35, 41, 48, 55 and 79 years.
  • the cell isolation is performed as described by Zonari et al. Briefly, the tissue samples are cut into small pieces of 0.5 cm2 and incubated in PBS containing dispase (2.5 U/mL, BD Biosciences) overnight at 4°C. The epidermis is then mechanically separated from the dermis and incubated in 0.5% trypsin-EDTA (Gibco, USA) for 7 min at 37°C to isolate the keratinocytes.
  • the cells are separated from the remaining tissue using a 100 mm pore size cell strainer (BD Biosciences, USA) and the cell suspension is centrifuged at 290 g for 5 min.
  • Human keratinocytes are seeded at a density of 100,000 cells/ cm in Keratinocyte Serum Free Medium (KSFM) supplemented with Epidermal growth factor and Bovine pituitary extract (Gibco).
  • KSFM Keratinocyte Serum Free Medium
  • Gibco Bovine pituitary extract
  • the dermis separated from the epidermis is incubated in PBS containing collagenase IA (250 U/mL, Sigma) for 3 hr at 37°C.
  • the HDF are separated from the remaining tissue using a 100 mm pore size cell strainer, centrifuged at 300 g for 5 min and seeded at a density of 50,000 cells/cm in Dulbecco's Modified Eagle Medium (Invitrogen, Carlsbad, CA),supplemented with 10% v.v. Fetal Bovine Serum (FBS; VWR) and 1% v.v. Penicillin-Streptomycin (Invitrogen).
  • FBS Fetal Bovine Serum
  • VWR Fetal Bovine Serum
  • Penicillin-Streptomycin Invitrogen.
  • Topical formulation 1 Pharmaceutical composition for topical application.
  • Topical formulation 2 Pharmaceutical composition for topical application.
  • mice are treated with compound selected from the table 1 ab and Table 1c by protocol selected from the Example 1.
  • COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c counteracts frailty and loss of renal function in naturally aged mice as well improves fur.
  • compound selected from the table 1 ab and Table 1c target senescence and tissue homeostasis in normal mice that are allowed to age naturally.
  • the biological variation in p16- driven senescence is substantial in aged p16 ⁇ 3MR, compared to young Xpd TTD/TTD -p16 ⁇ .
  • the variation in running wheel activity is too large to perform meaningful experiments.
  • COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c significantly reduces p16-driven RLUC, and improve fur density and responsiveness . Furthermore, in the kidneys of these mice COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c increases the number of LMNB1 positive cells, reduces IL-6 expression and restores renal filtering capacity measured by decreased plasma Urea. As an extra control, also the plasma levels of a second metabolite indicative of reduced renal function, Creatinine, can be measured.
  • COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c is improved by COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c , independently confirming the beneficial effect of COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c on the restoration of renal filtering capacity in naturally aged mice.
  • administration of COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c or GCV reduce plasma Urea and Creatinine levels.
  • senescent cells are causal for the reduction in renal function in fast aging Xpd TTD/TTD and naturally aged wildtype mice and by selective targeting of high-SASP expressing senescent cells in the tubuli, COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c can restore kidney homeostasis.
  • COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c may thus be a potent drug to restore loss of health after natural aging and is an attractive option to explore further in the battle against those age-related diseases that are at least in part driven by senescence.
  • COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c potently counteracted the Doxorubicin-induced increase in plasma AST.
  • mice are used for Doxorubicin-experiments at 10– 40wks of age, for Xpd TTD/TTD vs. wt experiments at 26–60 wks of age and for naturally aged mice at 115–130wks of age. All mice are kept in group housing until the start of the experiment after which they are placed in individual cages containing free access to a running wheel. Both sexes are used throughout the study. Where feasible, littermates of the same sex are used. These are randomly assigned to experimental groups.
  • Fur density analysis Xpd TTD/TTD mice show reduced fur density.
  • the phenotype is ranged from 0–4 where 0 is very patched and 4 is wildtype.
  • Each mouse is scored before and after the experiment. The final score is determined as the ratio (final- baseline)/baseline and the % change is subsequently plotted.
  • the experiment contains mice from two independent cohorts. For naturally aged mice only males can be included, since one often do not observe significant hair loss in females. In these cohorts, around 80% shows (varying degrees of) loss of hair at the beginning (115+wks), or developed it over the course of the experiment.
  • Plasma values as measure for tissue function On the indicated time points, whole blood samples are collected in a Microvette with Lithium Heparin (Sarstedt) for plasma separation and spun for 10min at 4.6 ⁇ g. The (clear) supernatant is transferred into regular 1.5ml tubes and spun again for 5min at 4.6 ⁇ g. The supernatants are transferred again into 1.5ml tubes, snap frozen in liquid N2 and stored at ⁇ 80°C. [AST] is measured using an AST Activity Assay Kit (Sigma). The samples are incubated with 100 ⁇ l reaction mix in a 96 well plate and place at 37°C. The absorbance at 450nm is determined after 2 minutes for baseline analysis and after 40 minutes for a final analysis.
  • [Urea] is measured using a QuantiChrom Urea Assay Kit (Gentaur). The samples are incubated in 200 ⁇ l reaction mix for 10 minutes at room temperature before absorbance is measured at 520nm.
  • [Creatinine] is measured using Creatinine Assay Kit (Sigma). Samples are incubated with 50 ⁇ l reaction mix at 37°C for 60 minutes and the absorbance is measured at 570nm. Ratios comparing plasma values after treatment compared to baseline are determined and plotted as % over baseline in scatter plots.
  • Kidney slice culture Freshly isolated kidneys are sectioned in 200 M thick slices using a Vibratome (Leica, Eindhoven, the Netherlands).
  • the sections are cultured in Dulbecco’s modified eagle medium with 10% FCS at 37 °C, 5% CO2 on a shaker (60 rpm). Following incubation with shRNA- containing lentiviral particles, or COMPOUND SELECTED FROM THE TABLE 1 ab and Table 1c , as indicated, the slices are fixed for 30min in formalin and stored at ⁇ 80. Subsequently, they are subsectioned to 10 M slices using a Cryostat, placed on a charged microscopy slide and processed for TUNEL positivity.
  • topical application of compound selected from the table 1 ab and Table 1c is effective in treatment of alopecia (In humans) and fur improvement (in mice and other animals).
  • application of compound selected from the table 1 ab and Table 1c is effective in treatment of alopecia (In humans if administered PO or IV) and fur improvement (in mice and other animals if administered PO or IV or IP).
  • application of compound selected from the table 1 ab and Table 1c is effective in treatment of alopecia (In humans if administered in dosage selected from Example 10 PO or IV) and fur improvement (in mice and other animals if administered in dosage selected from Example 1 PO or IV or IP )
  • More details on the protocol can be found in Baar MP, Brandt RMC, Putavet DA, et al. Targeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and Aging.
  • EXAMPLE 34 Lisuride Patch known in the art for transdermal delivery can be a non-limiting example for composition and delivery device for any one of the compounds selected from the table 1 ab and Table 1c .
  • EXAMPLE 35 Any Example of the treatment suggested by this application, wherein the treatment groups of mice is treated each – by the protocol suggested below until the at least one symptom of the respective disease is not alliviated. 1. Compound selected from the table 1 ab and Table 1c , PO administration, every 24h 0.001 mg/kg 2. Compound selected from the table 1 ab and Table 1c , PO administration, every 24h 0.005 mg/kg 3.

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Hospice & Palliative Care (AREA)
  • Ophthalmology & Optometry (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Psychiatry (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Psychology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
PCT/RU2022/050044 2021-02-10 2022-02-09 Composés, compositions et méthodes de traitement de maladies et d'états liés à l'âge WO2022173333A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP22753056.5A EP4291239A2 (fr) 2021-02-10 2022-02-09 Composés, compositions et méthodes de traitement de maladies et d'états liés à l'âge

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
RU2021103259 2021-02-10
RU2021103259 2021-02-10

Publications (1)

Publication Number Publication Date
WO2022173333A2 true WO2022173333A2 (fr) 2022-08-18

Family

ID=82838892

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/RU2022/050044 WO2022173333A2 (fr) 2021-02-10 2022-02-09 Composés, compositions et méthodes de traitement de maladies et d'états liés à l'âge

Country Status (2)

Country Link
EP (1) EP4291239A2 (fr)
WO (1) WO2022173333A2 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117198550A (zh) * 2023-11-07 2023-12-08 北京青颜博识健康管理有限公司 基于大数据的胶原蛋白肽抗衰老评估系统

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117198550A (zh) * 2023-11-07 2023-12-08 北京青颜博识健康管理有限公司 基于大数据的胶原蛋白肽抗衰老评估系统
CN117198550B (zh) * 2023-11-07 2024-03-19 北京青颜博识健康管理有限公司 基于大数据的胶原蛋白肽抗衰老评估系统

Also Published As

Publication number Publication date
EP4291239A2 (fr) 2023-12-20

Similar Documents

Publication Publication Date Title
US11963957B2 (en) Treating cardiovascular disease by selectively eliminating senescent cells
US20230218617A1 (en) Killing senescent cells and treating senescence-associated conditions using a src inhibitor and a flavonoid
EP4291239A2 (fr) Composés, compositions et méthodes de traitement de maladies et d'états liés à l'âge
JP6918839B2 (ja) 概日時計の乱れに関連するマイクロバイオームの調節異常を処置するための方法及び医薬組成物
US20200317640A1 (en) Acyl Sulfonamides that are BCL Family Antagonists for Use in Clinical Management of Conditions Caused or Mediated by Senescent Cells and for Treating Cancer
WO2022173334A2 (fr) Procédés, compositions et composés pour le traitement de maladies et d'états liés à l'âge
US20230190748A1 (en) Compositions for treatment of aged diseases
WO2023014718A2 (fr) 25-hydroxycholestérol (25 hc), inhibiteur d'agrégation de cryab pour améliorer la raideur vasculaire
WO2023230593A1 (fr) Inhibiteurs de ptp1b pour traitement d'une lésion pulmonaire
Devine Tumor Induced Cardiovascular Dysfunction

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22753056

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2022753056

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022753056

Country of ref document: EP

Effective date: 20230911